1
|
Yang W, Zhang Y, Ni Q, Huang AQ, Tang Y, Xu X, Zeng C. High-resolution LC-MS/MS combined with TMT quantitative proteomic analysis reveals regulatory mechanism of sperm capacitation by heparin, Ca 2+ and BSA. Int J Biol Macromol 2025; 305:141349. [PMID: 39986510 DOI: 10.1016/j.ijbiomac.2025.141349] [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: 11/24/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Heparin, BSA, and CaCl2 have been demonstrated to induce sperm capacitation in vitro; however, the specific molecular mechanisms by which different chemokines regulate sperm capacitation remain incompletely elucidated. In previous studies, our laboratory utilized various chemokines to induce capacitation in porcine sperm in vitro and classified these chemokines into three categories based on changes in olfactory receptors. Therefore, this study aims to systematically investigate the molecular pathways and regulatory mechanisms underlying heparin, CaCl2, and BSA induced porcine sperm capacitation. Porcine sperm were treated with heparin, CaCl2, or BSA for 1-4 h, and capacitation was assessed by measuring mitochondrial membrane potential (MMP), intracellular Ca2+ concentration, and capacitation rate. Differential protein expression among the three groups was analyzed using TMT-based quantitative proteomics. The results demonstrated that heparin, CaCl2, and BSA significantly increased intracellular Ca2+ concentration in a time-dependent manner, reduced MMP, and successfully induced sperm capacitation. Proteomic analysis revealed that differentially expressed proteins between the heparin and BSA groups were primarily enriched in lipid metabolism-related signaling pathways, such as PPAR and AMPK, while differentially expressed proteins in the CaCl2 group were significantly enriched in B vitamin metabolic pathways, including riboflavin and nicotinic acid metabolism. Furthermore, olfactory receptors OR1J4 and OR4C13 were found to specifically bind chemokines and participate in the regulation of sperm capacitation. In conclusion, this study elucidates the distinct molecular mechanisms by which heparin, CaCl2, and BSA induce porcine sperm capacitation and provides the first evidence of the critical role of olfactory receptors OR1J4 and OR4C13 in regulating sperm capacitation, offering new theoretical insights into the molecular mechanisms underlying sperm capacitation.
Collapse
Affiliation(s)
- Weihan Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Qingyong Ni
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - An-Qi Huang
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Yiguo Tang
- Daocheng Kangbaqing Animal Husbandry Co., Ltd, Sichuan, China
| | - Xinhong Xu
- Daocheng Kangbaqing Animal Husbandry Co., Ltd, Sichuan, China
| | - Changjun Zeng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China; College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China.
| |
Collapse
|
2
|
Adnane M, Ahmed M, Chapwanya A. Advances in Molecular Biology and Immunology of Spermatozoa and Fertilization in Domestic Animals: Implications for Infertility and Assisted Reproduction. Curr Mol Med 2025; 25:167-186. [PMID: 39572916 DOI: 10.2174/0115665240306965240802075331] [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: 01/27/2024] [Revised: 04/25/2024] [Accepted: 06/13/2024] [Indexed: 04/11/2025]
Abstract
Unlocking the secrets of reproductive success in domestic animals requires a deep understanding of the molecular biology and immunology of spermatozoa, capacitation, fertilization, and conception. This review highlights the complex processes involved in spermatogenesis and sperm capacitation, including changes in membrane properties, signaling pathways, and the crucial acrosome reaction. The interaction with the zona pellucida in species-specific gamete recognition and binding is emphasized. The implications of fertilization defects for infertility and assisted reproduction are discussed, underscoring the challenges faced in breeding programs. The future directions for research in this field involve advancements in molecular techniques, understanding the immune regulation of spermatozoa, investigating environmental factors' impact, and integrating multi-omics approaches to enhance assisted reproduction techniques in domestic animals. This review contributes to our understanding of the intricate mechanisms underlying successful reproduction and provides insights into potential strategies for improving fertility outcomes in domestic animals.
Collapse
Affiliation(s)
- Mounir Adnane
- Department of Biomedicine, Institute of Veterinary Sciences, University Ibn Khaldoun of Tiaret, Tiaret 14000, Algeria
| | - Moussa Ahmed
- Department of Animal Health, Institute of Veterinary Sciences, University Ibn Khaldoun of Tiaret, Tiaret, 14000, Algeria
| | - Aspinas Chapwanya
- Department of Clinical Sciences, Ross University School of Veterinary Medicine, Basseterre, 00265, Saint Kitts and Nevis
| |
Collapse
|
3
|
Kumar KN, Veerappa VG, Kumaresan A, Lavanya M, King JES, Sulochana M, Patil S, Jeyakumar S. Localization and expression analysis of sperm-specific glyceraldehyde 3-phosphate dehydrogenase in bull spermatozoa with contrasting sperm motility. Andrology 2024. [PMID: 39587844 DOI: 10.1111/andr.13810] [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: 05/20/2024] [Revised: 10/30/2024] [Accepted: 11/10/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Poor sperm motility leading to male infertility has become a profound crisis to be addressed in this contemporary era. In many cases, the origin of poor sperm motility remains unexplained. Few studies reported the indispensable role of sperm-specific glyceraldehyde 3-phosphate dehydrogenase (GAPDHS) in sperm motility, however, studies on GAPDHS are severely confined. OBJECTIVES The present study aimed to assess the localization patterns, expression levels, and enzyme activity of GAPDHS in normal and asthenozoospermic bulls and to examine their association with sperm functional parameters. MATERIALS AND METHODS The bull semen samples were classified into high-motile and low-motile groups (n = 7 per each group) based on the ejaculate rejection rate. Sperm kinetic parameters were assessed using computer-assisted sperm analysis (CASA). Sperm viability, mitochondrial membrane potential (MMP), reactive oxygen species (ROS), and intracellular calcium levels were measured through flow cytometry. Subsequently, GAPDHS localization was observed via immunocytochemistry. The expression levels and enzyme activity of GAPDHS were estimated using western blotting and a GAPDHS activity assay kit. RESULTS AND DISCUSSION Sperm viability, MMP, ROS, and live sperm intracellular calcium levels did not differ significantly between high and low motile groups. A significant positive correlation was found between MMP and sperm viability, whereas no significant association was found between MMP and sperm progressive motility. The GAPDHS was localized in the principal piece, head-midpiece junction, and at the acrosome region of bull sperm. GAPDHS localization intensity, expression levels, and enzyme activity were found significantly (p < 0.05) higher in the high motile group than in low motile group. Furthermore, we noticed a significant positive correlation between GAPDHS activity and sperm kinetic parameters. CONCLUSIONS The analysis of GAPDHS localization patterns, expression levels, and enzyme activity indicated its potential role in sperm motility, suggesting that GAPDHS could serve as a candidate biomarker for sperm motility and male fertility.
Collapse
Affiliation(s)
- K Naresh Kumar
- Semen Technology Laboratory, Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Adugodi, Bengaluru, India
| | - Vedamurthy G Veerappa
- Semen Technology Laboratory, Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Adugodi, Bengaluru, India
| | - Arumugam Kumaresan
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research, Institute, Adugodi, Bengaluru, India
| | - Maharajan Lavanya
- Semen Technology Laboratory, Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Adugodi, Bengaluru, India
| | - J Ebenezer Samuel King
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research, Institute, Adugodi, Bengaluru, India
| | - M Sulochana
- Semen Technology Laboratory, Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research Institute, Adugodi, Bengaluru, India
| | - Shivanagouda Patil
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research, Institute, Adugodi, Bengaluru, India
| | - Sakthivel Jeyakumar
- Dairy Production Section, Southern Regional Station of ICAR-National Dairy Research, Institute, Adugodi, Bengaluru, India
| |
Collapse
|
4
|
Ahmed Z, Liu M, Huang S, Xu D. CATSPER2 and SPEF2 are potential molecular markers for boar sperm quality: a population association study. J Assist Reprod Genet 2024; 41:3105-3117. [PMID: 39312031 PMCID: PMC11621296 DOI: 10.1007/s10815-024-03252-0] [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: 05/16/2024] [Accepted: 09/05/2024] [Indexed: 12/06/2024] Open
Abstract
PURPOSE This study investigates the role of cation channel sperm associated 2 (CATSPER2) and sperm flagella 2 (SPEF2) genes in boar spermatogenesis, focusing on their association with sperm quality traits in boars. METHODS Utilizing targeted next-generation sequencing, we identified and genotyped two polymorphisms in CATSPER2 (rs341636020G > A, rs326912346G > T) and three variants in SPEF2 (rs320839956A > G, rs334209514C > A, rs325319860C > T) across three boar breeds (Duroc, n = 181; Landrace, n = 87; Large White, n = 52). RESULTS Our results confirmed the presence of the specified single nucleotide polymorphisms (SNPs), adhering to association study criteria. In CATSPER2, significant associations were detected between rs341636020G > A and sperm curvilinear velocity (VCL) in Duroc and Landrace boars, and between rs326912346G > T and straight velocity (VSL) in Duroc and Large White boars. For SPEF2, rs320839956A > G was significantly linked to sperm viability in Duroc and Landrace and to sperm concentration (SCON) in Large White boars. Additionally, rs334209514C > A and rs325319860C > T showed significant associations with SCON and VCL respectively, in Doruc and Landrace boars. CONCLUSIONS Overall, our findings suggest that CATSPER2 and SPEF2 SNPs significantly impact boar sperm quality traits. These genetic markers have the potential to enhance boar fertility through selective breeding programs, contributing to the optimization of reproductive performance in pigs.
Collapse
Affiliation(s)
- Zulfiqar Ahmed
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, and Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, Hubie, People's Republic of China
| | - Min Liu
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, and Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, Hubie, People's Republic of China
| | - Shuntao Huang
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, and Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, Hubie, People's Republic of China
| | - Dequan Xu
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, and Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, Hubie, People's Republic of China.
| |
Collapse
|
5
|
Osycka-Salut CE, Waremkraut M, Garaguso R, Piga E, Martínez-León E, Marín-Briggiler CI, Gervasi MG, Navarro M, Visconti PE, Buffone MG, Mutto AA, Krapf D. Treatment of cryopreserved bovine sperm with calcium ionophore A23187 increases in vitro embryo production. Theriogenology 2024; 229:1-7. [PMID: 39133991 DOI: 10.1016/j.theriogenology.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024]
Abstract
After ejaculation, mammalian sperm undergo a series of molecular events conducive to the acquisition of fertilizing competence. These events are collectively known as capacitation and involve acrosomal responsiveness and a vigorous sperm motility called hyperactivation. When mimicked in the laboratory, capacitating bovine sperm medium contains bicarbonate, calcium, albumin and heparin, among other components. In this study, we aimed at establishing a new capacitation protocol for bovine sperm, using calcium ionophore. Similar to our findings using mouse sperm, bovine sperm treated with Ca2+ ionophore A23187 were quickly immobilized. However, these sperm initiated capacitation after ionophore removal in fresh medium without heparin, and independent of the Protein Kinase A. When A23187-treated sperm were used on in vitro fertilization (IVF) procedures without heparin, eggs showed cleavage rates similar to standardized IVF protocols using heparin containg synthetic oviduct fluid (IVF-SOF). However, when A23187 pre-treated sperm were further used for inseminating eggs in complete IVF-SOF-heparin, a significantly higher percentage of embryo development was observed, suggesting a synergism between two different signaling pathways during bovine sperm capacitation. These results have the potential to improve current protocols for bovine IVF that could also be applied in other species of commercial interest.
Collapse
Affiliation(s)
- C E Osycka-Salut
- Instituto de Investigaciones Biotecnológicas (IIBio-UNSAM-CONICET), Buenos Aires C1650, Argentina
| | - M Waremkraut
- Instituto de Investigaciones Biotecnológicas (IIBio-UNSAM-CONICET), Buenos Aires C1650, Argentina
| | - R Garaguso
- Instituto de Investigaciones Biotecnológicas (IIBio-UNSAM-CONICET), Buenos Aires C1650, Argentina
| | - E Piga
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET-UNR), Rosario, Santa Fe S2000EZP, Argentina
| | - E Martínez-León
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", CABA, 1120, Argentina
| | - C I Marín-Briggiler
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428ADN, Argentina
| | - M G Gervasi
- Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| | - M Navarro
- Instituto de Investigaciones Biotecnológicas (IIBio-UNSAM-CONICET), Buenos Aires C1650, Argentina
| | - P E Visconti
- Department of Veterinary and Animal Science, Paige Labs, University of Massachusetts, Amherst, MA 01003, USA
| | - M G Buffone
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428ADN, Argentina
| | - A A Mutto
- Instituto de Investigaciones Biotecnológicas (IIBio-UNSAM-CONICET), Buenos Aires C1650, Argentina.
| | - D Krapf
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET-UNR), Rosario, Santa Fe S2000EZP, Argentina.
| |
Collapse
|
6
|
Novero AG, Torres Rodríguez P, De la Vega Beltrán JL, Schiavi-Ehrenhaus LJ, Luque GM, Carruba M, Stival C, Gentile I, Ritagliati C, Santi CM, Nishigaki T, Krapf D, Buffone MG, Darszon A, Treviño CL, Krapf D. The sodium-proton exchangers sNHE and NHE1 control plasma membrane hyperpolarization in mouse sperm. J Biol Chem 2024; 300:107932. [PMID: 39476963 PMCID: PMC11629550 DOI: 10.1016/j.jbc.2024.107932] [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: 07/30/2024] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 11/06/2024] Open
Abstract
Sperm capacitation is a complex process that takes place in the female reproductive tract and empowers mammalian sperm with the competence to fertilize an egg. It consists of an intricate cascade of events that can be mimicked in vitro through incubation in a medium containing essential components, such as bicarbonate, albumin, Ca2+, and energy substrates, among others. Genetic and pharmacological studies have underscored the unique significance of the K+ channel SLO3 in membrane potential hyperpolarization, as evidenced by the infertility of mice lacking its expression. Notably, two key molecular events, sperm hyperpolarization and intracellular alkalinization, are central to the capacitation process. SLO3 is activated by alkalinization. However, the molecular mechanisms responsible for intracellular alkalization and activation of SLO3 are not completely understood. In this study, we examined the impact of Na+/H+ exchangers (NHEs) on mouse sperm membrane hyperpolarization during capacitation. Pharmacological inhibition of the NHE1 blocked membrane hyperpolarization. A similar effect was observed in sperm deficient of the Ca2+ channel CatSper because of NHE1 not being activated by Ca2+. In addition, the sperm-specific NHE (sNHE) KO did not show membrane hyperpolarization upon capacitation or induction with cAMP analogs. Our results show that sNHE is dually modulated by cAMP and membrane hyperpolarization probably through its cyclic nucleotide-binding domain and the voltage-sensor motif, respectively. Together, sNHE and NHE1 provide the alkalinization need for SLO3 activation during capacitation.
Collapse
Affiliation(s)
- Analia G Novero
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario, Argentina
| | | | | | - Liza J Schiavi-Ehrenhaus
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Guillermina M Luque
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Micaela Carruba
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario, Argentina
| | - Cintia Stival
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario, Argentina
| | - Iñaki Gentile
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario, Argentina
| | - Carla Ritagliati
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario, Argentina
| | - Celia M Santi
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, Missouri, USA
| | | | - Diego Krapf
- Department of Electrical and Computer Engineering, Colorado State University, Fort Collins, Colorado, USA
| | - Mariano G Buffone
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | | | | | - Dario Krapf
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario, Argentina.
| |
Collapse
|
7
|
Niu H, Zhou M, Ji A, Zogona D, Wu T, Xu X. Molecular Mechanism of Pasteurized Akkermansia muciniphila in Alleviating Type 2 Diabetes Symptoms. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:13083-13098. [PMID: 38829529 DOI: 10.1021/acs.jafc.4c01188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Type 2 diabetes (T2DM) significantly diminishes people's quality of life and imposes a substantial economic burden. This pathological progression is intimately linked with specific gut microbiota, such as Akkermansia muciniphila. Pasteurized A. muciniphila (P-AKK) has been defined as a novel food by the European Food Safety Authority and exhibited significant hypoglycemic activity. However, current research on the hypoglycemic activity of P-AKK is limited to the metabolic level, neglecting systematic exploration at the pathological level. Consequently, its material basis and mechanism of action for hypoglycemia remain unclear. Drawing upon this foundation, we utilized high-temperature killed A. muciniphila (H-K-AKK) with insignificant hypoglycemic activity as the control research object. Assessments were conducted at pathological levels to evaluate the hypoglycemic functions of both P-AKK and H-K-AKK separately. Our study unveiled for the first time that P-AKK ameliorated symptoms of T2DM by enhancing the generation of glucagon-Like Peptide 1 (GLP-1), with pasteurized A. muciniphila total proteins (PP) being a pivotal component responsible for this activity. Utilizing SDS-PAGE, proteomics, and molecular docking techniques, we deeply analyzed the material foundation of PP. We scientifically screened and identified a protein weighing 77.85 kDa, designated as P5. P5 enhanced GLP-1 synthesis and secretion by activating the G protein-coupled receptor (GPCR) signaling pathway, with free fatty acid receptor 2 (FFAR-2) being identified as the pivotal target protein for P5's physiological activity. These findings further promote the widespread application of P-AKK in the food industry, laying a solid theoretical foundation for its utilization as a beneficial food ingredient or functional component.
Collapse
Affiliation(s)
- Huifang Niu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit Vegetable Processing Quality Control (Huazhong Agricultural University), School of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Minfeng Zhou
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Anying Ji
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit Vegetable Processing Quality Control (Huazhong Agricultural University), School of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Daniel Zogona
- Department of Food & Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Ting Wu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit Vegetable Processing Quality Control (Huazhong Agricultural University), School of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Xiaoyun Xu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit Vegetable Processing Quality Control (Huazhong Agricultural University), School of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| |
Collapse
|
8
|
Lee KH, Hwang JY. Ca 2+ homeostasis and male fertility: a target for a new male contraceptive system. Anim Cells Syst (Seoul) 2024; 28:171-183. [PMID: 38686363 PMCID: PMC11057403 DOI: 10.1080/19768354.2024.2345647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024] Open
Abstract
Ca2+ is a key secondary messenger that determines sperm motility patterns. Mammalian sperm undergo capacitation, a process to acquire fertilizing ability, in the female reproductive tract. Capacitated sperm change their flagellar waveform to develop hyperactivated motility, which is crucial for successful sperm navigation to the eggs and fertilization. The sperm-specific channel, CATSPER, and an ATPase transporter, PMCA4, serve as major paths for Ca2+ influx and efflux, respectively, in sperm. The ionic paths coordinate Ca2+ homeostasis in the sperm, and their loss-of-function impairs sperm motility, to cause male infertility. In this review, we summarize the physiological significance of these two Ca2+ gates and suggest their potential applications in novel male contraceptives.
Collapse
Affiliation(s)
- Kyung-Ha Lee
- Department of Molecular Biology, Pusan National University, Busan, South Korea
- Institute of Systems Biology, Pusan National University, Busan, South Korea
| | - Jae Yeon Hwang
- Department of Molecular Biology, Pusan National University, Busan, South Korea
- Institute of Systems Biology, Pusan National University, Busan, South Korea
| |
Collapse
|
9
|
Zhang X, Liang M, Song D, Huang R, Chen C, Liu X, Chen H, Wang Q, Sun X, Song J, Zhang J, Kang H, Zeng X. Both protein and non-protein components in extracellular vesicles of human seminal plasma improve human sperm function via CatSper-mediated calcium signaling. Hum Reprod 2024; 39:658-673. [PMID: 38335261 DOI: 10.1093/humrep/deae018] [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: 10/08/2023] [Revised: 01/10/2024] [Indexed: 02/12/2024] Open
Abstract
STUDY QUESTION What is the significance and mechanism of human seminal plasma extracellular vesicles (EVs) in regulating human sperm functions? SUMMARY ANSWER EV increases the intracellular Ca2+ concentrations [Ca2+]i via extracellular Ca2+ influx by activating CatSper channels, and subsequently modulate human sperm motility, especially hyperactivated motility, which is attributed to both protein and non-protein components in EV. WHAT IS KNOWN ALREADY EVs are functional regulators of human sperm function, and EV cargoes from normal and asthenozoospermic seminal plasma are different. Pre-fusion of EV with sperm in the acidic and non-physiological sucrose buffer solution could elevate [Ca2+]i in human sperm. CatSper, a principle Ca2+ channel in human sperm, is responsible for the [Ca2+]i regulation when sperm respond to diverse extracellular stimuli. However, the role of CatSper in EV-evoked calcium signaling and its potential physiological significance remain unclear. STUDY DESIGN, SIZE, DURATION EV isolated from the seminal plasma of normal and asthenozoospermic semen were utilized to investigate the mechanism by which EV regulates calcium signal in human sperm, including the involvement of CatSper and the responsible cargoes in EV. In addition, the clinical application potential of EV and EV protein-derived peptides were also evaluated. This is a laboratory study that went on for more than 5 years and involved more than 200 separate experiments. PARTICIPANTS/MATERIALS, SETTING, METHODS Semen donors were recruited in accordance with the Institutional Ethics Committee on human subjects of the Affiliated Hospital of Nantong University and Jiangxi Maternal and Child Health Hospital. The Flow NanoAnalyzer, western blotting, and transmission electron microscope were used to systematically characterize seminal plasma EV. Sperm [Ca2+]i responses were examined by fluorimetric measurement. The whole-cell patch-clamp technique was performed to record CatSper currents. Sperm motility parameters were assessed by computer-assisted sperm analysis. Sperm hyperactivation was also evaluated by examining their penetration ability in viscous methylcellulose media. Protein and non-protein components in EV were analyzed by liquid chromatography-mass spectrum. The levels of prostaglandins, reactive oxygen species, malonaldehyde, and DNA integrity were detected by commercial kits. MAIN RESULTS AND THE ROLE OF CHANCE EV increased [Ca2+]i via an extracellular Ca2+ influx, which could be suppressed by a CatSper inhibitor. Also, EV potentiated CatSper currents in human sperm. Furthermore, the EV-in [Ca2+]i increase and CatSper currents were absent in a CatSper-deficient sperm, confirming the crucial role of CatSper in EV induced Ca2+ signaling in human sperm. Both proteins and non-protein components of EV contributed to the increase of [Ca2+]i, which were important for the effects of EV on human sperm. Consequently, EV and its cargos promoted sperm hyperactivated motility. In addition, seminal plasma EV protein-derived peptides, such as NAT1-derived peptide (N-P) and THBS-1-derived peptide (T-P), could activate the sperm calcium signal and enhance sperm function. Interestingly, EV derived from asthenozoospermic semen caused a lower increase of [Ca2+]i than that isolated from normal seminal plasma (N-EV), and N-EV significantly improved sperm motility and function in both asthenozoospermic samples and frozen-thawed sperm. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This was an in vitro study and caution must be taken when extrapolating the physiological relevance to in vivo regulation of sperm. WIDER IMPLICATIONS OF THE FINDINGS Our findings demonstrate that the CatSper-mediated-Ca2+ signaling is involved in EV-modulated sperm function under near physiological conditions, and EV and their derivates are a novel CatSper and sperm function regulators with potential for clinical application. They may be developed to improve sperm motility resulting from low [Ca2+]i response and/or freezing and thawing. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by the National Natural Science Foundation of China (32271167), the Social Development Project of Jiangsu Province (BE2022765), the Nantong Social and People's Livelihood Science and Technology Plan (MS22022087), the Basic Science Research Program of Nantong (JC22022086), and the Jiangsu Innovation and Entrepreneurship Talent Plan (JSSCRC2021543). The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Xiaoning Zhang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Min Liang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Dandan Song
- Institute of Life Science and School of Life Science, Nanchang University, Nanchang, China
| | - Rongzu Huang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Chen Chen
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Xiaojun Liu
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Houyang Chen
- Reproductive Medical Center, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Qingxin Wang
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoli Sun
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Jian Song
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiali Zhang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Hang Kang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Xuhui Zeng
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| |
Collapse
|
10
|
Novero AG, Rodríguez PT, De la Vega Beltrán JL, Schiavi-Ehrenhaus LJ, Luque GM, Carruba M, Stival C, Gentile I, Ritagliati C, Santi CM, Nishigaki T, Krapf D, Buffone MG, Darszon A, Treviño CL, Krapf D. The sodium-proton exchangers sNHE and NHE1 control plasma membrane hyperpolarization in mouse sperm. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583310. [PMID: 38496535 PMCID: PMC10942401 DOI: 10.1101/2024.03.04.583310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Sperm capacitation, crucial for fertilization, occurs in the female reproductive tract and can be replicated in vitro using a medium rich in bicarbonate, calcium, and albumin. These components trigger the cAMP-PKA signaling cascade, proposed to promote hyperpolarization of the mouse sperm plasma membrane through activation of SLO3 K+ channel. Hyperpolarization is a hallmark of capacitation: proper membrane hyperpolarization renders higher in vitro fertilizing ability, while Slo3 KO mice are infertile. However, the precise regulation of SLO3 opening remains elusive. Our study challenges the involvement of PKA in this event and reveals the role of Na+/H+ exchangers. During capacitation, calcium increase through CatSper channels activates NHE1, while cAMP directly stimulates the sperm-specific NHE, collectively promoting the alkalinization threshold needed for SLO3 opening. Hyperpolarization then feeds back Na+/H+ activity. Our work is supported by pharmacology, and a plethora of KO mouse models, and proposes a novel pathway leading to hyperpolarization.
Collapse
Affiliation(s)
- Analia G Novero
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario SF2000, Argentina
| | | | | | - Liz J Schiavi-Ehrenhaus
- Instituto de Bíologia y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Guillermina M Luque
- Instituto de Bíologia y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Micaela Carruba
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario SF2000, Argentina
| | - Cintia Stival
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario SF2000, Argentina
| | - Iñaki Gentile
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario SF2000, Argentina
| | - Carla Ritagliati
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario SF2000, Argentina
| | - Celia M Santi
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis Missouri 63110, USA
| | | | - Diego Krapf
- Department of Electrical and Computer Engineering, Colorado State University, Fort Collins, CO 80526, USA
| | - Mariano G Buffone
- Instituto de Bíologia y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | | | | | - Dario Krapf
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, and Laboratorio de Medicina Reproductiva, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Rosario SF2000, Argentina
| |
Collapse
|
11
|
Cavarocchi E, Sayou C, Lorès P, Cazin C, Stouvenel L, El Khouri E, Coutton C, Kherraf ZE, Patrat C, Govin J, Thierry-Mieg N, Whitfield M, Ray PF, Dulioust E, Touré A. Identification of IQCH as a calmodulin-associated protein required for sperm motility in humans. iScience 2023; 26:107354. [PMID: 37520705 PMCID: PMC10382937 DOI: 10.1016/j.isci.2023.107354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/23/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023] Open
Abstract
Sperm fertilization ability mainly relies on proper sperm progression through the female genital tract and capacitation, which involves phosphorylation signaling pathways triggered by calcium and bicarbonate. We performed exome sequencing of an infertile asthenozoospermic patient and identified truncating variants in MAP7D3, encoding a microtubule-associated protein, and IQCH, encoding a protein of unknown function with enzymatic and signaling features. We demonstrate the deleterious impact of both variants on sperm transcripts and proteins from the patient. We show that, in vitro, patient spermatozoa could not induce the phosphorylation cascades associated with capacitation. We also provide evidence for IQCH association with calmodulin, a well-established calcium-binding protein that regulates the calmodulin kinase. Notably, we describe IQCH spatial distribution around the sperm axoneme, supporting its function within flagella. Overall, our work highlights the cumulative pathological impact of gene mutations and identifies IQCH as a key protein required for sperm motility and capacitation.
Collapse
Affiliation(s)
- Emma Cavarocchi
- Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Camille Sayou
- Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Patrick Lorès
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, 75014 Paris, France
| | - Caroline Cazin
- Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France
- CHU de Grenoble Alpes, UM GI-DPI, 38000 Grenoble, France
| | - Laurence Stouvenel
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, 75014 Paris, France
| | - Elma El Khouri
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, 75014 Paris, France
| | - Charles Coutton
- Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France
- CHU Grenoble Alpes, UM de Génétique Chromosomique, Grenoble, France
| | | | - Catherine Patrat
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, 75014 Paris, France
- Laboratoire d’Histologie Embryologie - Biologie de la Reproduction - CECOS Groupe Hospitalier Universitaire Paris Centre, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - Jérôme Govin
- Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France
| | | | - Marjorie Whitfield
- Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Pierre F. Ray
- Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France
- CHU de Grenoble Alpes, UM GI-DPI, 38000 Grenoble, France
| | - Emmanuel Dulioust
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, 75014 Paris, France
- Laboratoire d’Histologie Embryologie - Biologie de la Reproduction - CECOS Groupe Hospitalier Universitaire Paris Centre, Assistance Publique-Hôpitaux de Paris, 75014 Paris, France
| | - Aminata Touré
- Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
12
|
Romarowski A, Fejzo J, Nayyab S, Martin-Hidalgo D, Gervasi MG, Balbach M, Violante S, Salicioni AM, Cross J, Levin LR, Buck J, Visconti PE. Mouse sperm energy restriction and recovery (SER) revealed novel metabolic pathways. Front Cell Dev Biol 2023; 11:1234221. [PMID: 37655160 PMCID: PMC10466171 DOI: 10.3389/fcell.2023.1234221] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 08/02/2023] [Indexed: 09/02/2023] Open
Abstract
Mammalian sperm must undergo capacitation to become fertilization-competent. While working on mice, we recently developed a new methodology for treating sperm in vitro, which results in higher rates of fertilization and embryo development after in vitro fertilization. Sperm incubated in media devoid of nutrients lose motility, although they remain viable. Upon re-adding energy substrates, sperm resume motility and become capacitated with improved functionality. Here, we explore how sperm energy restriction and recovery (SER) treatment affects sperm metabolism and capacitation-associated signaling. Using extracellular flux analysis and metabolite profiling and tracing via nuclear magnetic resonance (NMR) and mass spectrometry (MS), we found that the levels of many metabolites were altered during the starvation phase of SER. Of particular interest, two metabolites, AMP and L-carnitine, were significantly increased in energy-restricted sperm. Upon re-addition of glucose and initiation of capacitation, most metabolite levels recovered and closely mimic the levels observed in capacitating sperm that have not undergone starvation. In both control and SER-treated sperm, incubation under capacitating conditions upregulated glycolysis and oxidative phosphorylation. However, ATP levels were diminished, presumably reflecting the increased energy consumption during capacitation. Flux data following the fate of 13C glucose indicate that, similar to other cells with high glucose consumption rates, pyruvate is converted into 13C-lactate and, with lower efficiency, into 13C-acetate, which are then released into the incubation media. Furthermore, our metabolic flux data show that exogenously supplied glucose is converted into citrate, providing evidence that in sperm cells, as in somatic cells, glycolytic products can be converted into Krebs cycle metabolites.
Collapse
Affiliation(s)
- Ana Romarowski
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (IBYME-CONICET), Buenos Aires, Argentina
| | - Jasna Fejzo
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, United States
| | - Saman Nayyab
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | | | - Maria G. Gervasi
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Melanie Balbach
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Sara Violante
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ana M. Salicioni
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Justin Cross
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Lonny R. Levin
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Pablo E. Visconti
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
13
|
Rodríguez-Díaz R, Blanes-Zamora R, Vaca-Sánchez R, Gómez-Rodríguez J, Hardisson A, González-Weller D, Gutiérrez ÁJ, Paz S, Rubio C, González-Dávila E. Influence of Seminal Metals on Assisted Reproduction Outcome. Biol Trace Elem Res 2023; 201:1120-1134. [PMID: 35543968 PMCID: PMC9898355 DOI: 10.1007/s12011-022-03256-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023]
Abstract
Increased levels of metal ions in human seminal fluid have a significant correlation with male fertility. Few publications explain the effect of metals in semen and their influence on assisted reproductive treatments. Semen parameters and the levels of twenty-two metals were measured in the seminal fluid of 102 men attended in a Reproductive Unit. Metals were determined by optical emission spectrophotometry. A statistical relationship was found between spermiogram and iron, which was lower than expected in pathological spermiograms (p = 0.032); zinc (p = 0.066), calcium (p = 0.047), and magnesium (p = 0.048) mean levels were higher in normozoospermics. More days of sexual abstinence correlates with higher seminal zinc (p = 0.001) and magnesium levels (p = 0.002). Lower vanadium values were found to be associated with higher fertilization rates (p = 0.039). Higher values of lead (p = 0.052) and vanadium (p = 0.032) were obtained in patients who did not reach 100% embryo cleavage rate. Aluminium (p = 0.042) and sodium (p = 0.002) were found in lower amounts associated with better blastocyst rates. The implantation rate shows an inverse association with women's age and iron and calcium content, compared to magnesium and sodium which presented a significant direct association with this percentage. A significant direct relationship was found between the positive evolution of pregnancy and the values of zinc (p = 0.004), calcium (p = 0.013), potassium (p = 0.002), and magnesium (p = 0.009). The study confirms that zinc, iron, calcium, sodium, aluminium, magnesium, vanadium, and lead have positive-negative effects on reproduction and support the analysis of metals in semen as a new line of study on male fertility with implications for reproductive outcomes.
Collapse
Affiliation(s)
- Rubí Rodríguez-Díaz
- Human Reproduction Unit, Canary Islands University Hospital, San Cristobal de La Laguna, Spain.
- Obstetrics and Gynecology, University of La Laguna, San Cristobal de La Laguna, Tenerife, Spain.
| | - Raquel Blanes-Zamora
- Human Reproduction Unit, Canary Islands University Hospital, San Cristobal de La Laguna, Spain
- Obstetrics and Gynecology, University of La Laguna, San Cristobal de La Laguna, Tenerife, Spain
| | - Rebeca Vaca-Sánchez
- Human Reproduction Unit, Canary Islands University Hospital, San Cristobal de La Laguna, Spain
- Obstetrics and Gynecology, University of La Laguna, San Cristobal de La Laguna, Tenerife, Spain
| | - Jorge Gómez-Rodríguez
- Human Reproduction Unit, Canary Islands University Hospital, San Cristobal de La Laguna, Spain
- Obstetrics and Gynecology, University of La Laguna, San Cristobal de La Laguna, Tenerife, Spain
| | - Arturo Hardisson
- Toxicology, University of La Laguna, San Cristobal de La Laguna, Tenerife, Spain
| | | | - Ángel J Gutiérrez
- Toxicology, University of La Laguna, San Cristobal de La Laguna, Tenerife, Spain
| | - Soraya Paz
- Toxicology, University of La Laguna, San Cristobal de La Laguna, Tenerife, Spain
| | - Carmen Rubio
- Toxicology, University of La Laguna, San Cristobal de La Laguna, Tenerife, Spain
| | - E González-Dávila
- Mathematics, Statistics and Operations Research, University of La Laguna, San Cristobal de La Laguna, Tenerife, Spain
| |
Collapse
|
14
|
Constantinides M, Fayd’herbe De Maudave A, Potier-Cartereau M, Campos-Mora M, Cartron G, Villalba M. Direct Cell Death Induced by CD20 Monoclonal Antibodies on B Cell Lymphoma Cells Revealed by New Protocols of Analysis. Cancers (Basel) 2023; 15:cancers15041109. [PMID: 36831451 PMCID: PMC9954594 DOI: 10.3390/cancers15041109] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
CD20 monoclonal antibodies (mAbs) eliminate B cells in several clinical contexts. At least two of these Abs, obinutuzumab (OBI) and rituximab (RTX), induce quick elimination of targets and put cancer patients at risk of tumor lysis syndrome (TLS) within 12-24 h of the first dose. The mechanisms of killing can require the recruiting of effector mechanisms from the patient's immune system, but they can induce direct killing as well. This can be more rapid than recruiting cellular effectors and/or complement. We showed here that OBI and RTX induce quick (<1 h) and high (up to 60% for OBI) killing of two different B cell lines. This was unveiled by using two different techniques that circumvent cell centrifugation steps: a Muse® Cell Analyzer-based approach and a direct examination of the cells' physical properties by using forward scatter (FS) area and side scatter (SS) area by flow cytometry. These results excluded the presence of aggregates and were also confirmed by developing a normalized survival ratio based on the co-incubation of RTX- and OBI-sensitive cells with MOLM-13, an insensitive cell line. Finally, this normalized survival ratio protocol confirmed the RTX- and OBI-direct killing on primary tumor B cells from B cell chronic lymphocytic leukemia (B-CLL) and Non-Hodgkin's lymphoma (NHL) patients. Moreover, we unveiled that direct killing is higher than previously expected and absent in patients' samples at relapse. We also observed that these mAbs, prior to increasing intracellular calcium levels, decrease calcium entry, although manipulating calcium levels did not affect their cytotoxicity. Altogether, our results show that direct killing is a major mechanism to induce cell death by RTX and OBI mAbs.
Collapse
Affiliation(s)
- Michael Constantinides
- IRMB, University of Montpellier, INSERM, CHRU de Montpellier, F-34090 Montpellier, France
- Département d’Hématologie Clinique, CHRU de Montpellier, F-34090 Montpellier, France
| | | | - Marie Potier-Cartereau
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032 Tours, France; Réseau 3MC “Molécules Marines, Métabolisme et Cancer” and Réseau CASTOR “Cancers des Tissus Hormono-Dépendants” Cancéropôle Grand Ouest, F-34090 Montpellier, France
| | - Mauricio Campos-Mora
- IRMB, University of Montpellier, INSERM, CHRU de Montpellier, F-34090 Montpellier, France
| | - Guillaume Cartron
- Département d’Hématologie Clinique, CHRU de Montpellier, F-34090 Montpellier, France
| | - Martin Villalba
- IRMB, University of Montpellier, INSERM, CHRU de Montpellier, F-34090 Montpellier, France
- IRMB, University of Montpellier, INSERM, CNRS, CHRU de Montpellier, F-34090 Montpellier, France
- Correspondence:
| |
Collapse
|
15
|
The stallion sperm acrosome: Considerations from a research and clinical perspective. Theriogenology 2023; 196:121-149. [PMID: 36413868 DOI: 10.1016/j.theriogenology.2022.11.012] [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: 08/30/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022]
Abstract
During the fertilization process, the interaction between the sperm and the oocyte is mediated by a process known as acrosomal exocytosis (AE). Although the role of the sperm acrosome on fertilization has been studied extensively over the last 70 years, little is known about the molecular mechanisms that govern acrosomal function, particularly in species other than mice or humans. Even though subfertility due to acrosomal dysfunction is less common in large animals than in humans, the evaluation of sperm acrosomal function should be considered not only as a complementary but a routine test when individuals are selected for breeding potential. This certainly holds true for stallions, which might display lower levels of fertility in the face of "acceptable" sperm quality parameters determined by conventional sperm assays. Nowadays, the use of high throughput technologies such as flow cytometry or mass spectrometry-based proteomic analysis is commonplace in the research arena. Such techniques can also be implemented in clinical scenarios of males with "idiopathic" subfertility. The current review focuses on the sperm acrosome, with particular emphasis on the stallion. We aim to describe the physiological events that lead to the acrosome formation within the testis, the role of very specific acrosomal proteins during AE, the methods to study the occurrence of AE under in vitro conditions, and the potential use of molecular biology techniques to discover new markers of acrosomal function and subfertility associated with acrosomal dysfunction in stallions.
Collapse
|
16
|
Martin-Hidalgo D, González-Fernández L, Bragado MJ, Garcia-Marin LJ, Alves MG, Oliveira PF. The sirtuin 1 activator YK 3-237 stimulates capacitation-related events in human spermatozoa. Reprod Biomed Online 2023; 46:165-178. [PMID: 36357302 DOI: 10.1016/j.rbmo.2022.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/07/2022] [Accepted: 07/14/2022] [Indexed: 01/31/2023]
Abstract
RESEARCH QUESTION Does sirtuin-1 (SIRT1) have a role in the human spermatozoa capacitation process? DESIGN Human spermatozoa were incubated for 6 h in a capacitating medium in presence or absence of the specific SIRT1 activator, YK 3-237. Several sperm parameters were determined by flow cytometry: viability, acrosome reaction and mitochondria membrane status. Sperm motility was determined objectively by computer-assisted semen analysis. Sperm capacitation status was evaluated by the extent of protein tyrosine phosphorylation and by the percentage of spermatozoa with the acrosome reacted by a calcium ionophore challenge. RESULTS SIRT1 was detected in the connecting piece of human spermatozoa where a lysine acetylation pattern was mainly found along the sperm tail. SIRT1 activation accelerates the occurrence of a phenotype associated with human sperm capacitation, with no differences seen in the lysine acetylation pattern. After 1 h of co-incubation of YK 3-237 with human spermatozoa, tyrosine phosphorylation levels were comparable to control levels after 6 h of incubation in capacitating conditions. In addition, the activator improved sperm responsiveness to a Ca2+ ionophore (A23187) challenge determined by an increase in acrosome-reacted spermatozoa (P = 0.025). Importantly, sperm viability and mitochondrial activity-related parameters assessed by flow cytometry were not affected by YK 3-237. CONCLUSION YK 3-237 induces capacitation-related events in human spermatozoa such an increase of tyrosine phosphorylation levels and acrosome-reacted spermatozoa after the ionophore challenge. Together, these results show that YK 3-237 affects human spermatozoa capacitation-related events by a mechanism independent of protein lysine acetylation but dependent on bicarbonate and calcium.
Collapse
Affiliation(s)
- David Martin-Hidalgo
- Grupo de Investigación Señalización Intracelular y Tecnología de la Reproducción (SINTREP), Instituto de Investigación INBIO G+C, Universidad de Extremadura, Avda de la Universidad s/n, Cáceres 10003, Spain; Unit for Multidisciplinary Research in Biomedicine (UMIB), Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; Unidad de Investigación, Complejo Hospitalario Universitario de Cáceres, Avenida Pablo Naranjo s/n, Cáceres 10003, Spain.
| | - Lauro González-Fernández
- Grupo de Investigación Señalización Intracelular y Tecnología de la Reproducción (SINTREP), Instituto de Investigación INBIO G+C, Universidad de Extremadura, Avda de la Universidad s/n, Cáceres 10003, Spain
| | - M Julia Bragado
- Grupo de Investigación Señalización Intracelular y Tecnología de la Reproducción (SINTREP), Instituto de Investigación INBIO G+C, Universidad de Extremadura, Avda de la Universidad s/n, Cáceres 10003, Spain
| | - Luis J Garcia-Marin
- Grupo de Investigación Señalización Intracelular y Tecnología de la Reproducción (SINTREP), Instituto de Investigación INBIO G+C, Universidad de Extremadura, Avda de la Universidad s/n, Cáceres 10003, Spain
| | - Marco G Alves
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Pedro F Oliveira
- 7QOPNA and LAQV, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
17
|
Dai J, Li Q, Zhou Q, Zhang S, Chen J, Wang Y, Guo J, Gu Y, Gong F, Tan Y, Lu G, Zheng W, Lin G. IQCN disruption causes fertilization failure and male infertility due to manchette assembly defect. EMBO Mol Med 2022; 14:e16501. [PMID: 36321563 PMCID: PMC9728048 DOI: 10.15252/emmm.202216501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Total fertilization failure (TFF) is an important cause of infertility; however, the genetic basis of TFF caused by male factors remains to be clarified. In this study, whole-exome sequencing was firstly used to screen for genetic causes of TFF after intracytoplasmic sperm injection (ICSI), and homozygous variants in the novel gene IQ motif-containing N (IQCN) were identified in two affected individuals with abnormal acrosome structures. Then, Iqcn-knockout mice were generated by CRISPR-Cas9 technology and showed that the knockout male mice resembled the human phenotypes. Additionally, we found that IQCN regulates microtubule nucleation during manchette assembly via calmodulin and related calmodulin-binding proteins, which resulted in head deformity with aberrant oocyte activation factor PLCζ. Fortunately, ICSI with assisted oocyte activation can overcome IQCN-associate TFF and male infertility. Thus, our study firstly identified the function of IQCN, highlights the relationship between the manchette assembly and fertilization, and provides a genetic marker and a therapeutic option for male-source TFF.
Collapse
Affiliation(s)
- Jing Dai
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical ScienceCentral South UniversityChangShaChina,Reproductive and Genetic Hospital of CITIC‐XIANGYAChangShaChina,Clinical Research Center for Reproduction and Genetics in Hunan ProvinceChangShaChina
| | - Qi Li
- Reproductive Medicine Center, Xiangya HospitalCentral South UniversityChangShaChina
| | - Qinwei Zhou
- Reproductive and Genetic Hospital of CITIC‐XIANGYAChangShaChina,Clinical Research Center for Reproduction and Genetics in Hunan ProvinceChangShaChina
| | - Shen Zhang
- Reproductive and Genetic Hospital of CITIC‐XIANGYAChangShaChina,Clinical Research Center for Reproduction and Genetics in Hunan ProvinceChangShaChina
| | - Junru Chen
- Reproductive and Genetic Hospital of CITIC‐XIANGYAChangShaChina,Clinical Research Center for Reproduction and Genetics in Hunan ProvinceChangShaChina
| | - Yize Wang
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical ScienceCentral South UniversityChangShaChina
| | - Jing Guo
- Reproductive and Genetic Hospital of CITIC‐XIANGYAChangShaChina,Clinical Research Center for Reproduction and Genetics in Hunan ProvinceChangShaChina
| | - Yifan Gu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical ScienceCentral South UniversityChangShaChina,Reproductive and Genetic Hospital of CITIC‐XIANGYAChangShaChina,Clinical Research Center for Reproduction and Genetics in Hunan ProvinceChangShaChina,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning CommissionChangShaChina
| | - Fei Gong
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical ScienceCentral South UniversityChangShaChina,Reproductive and Genetic Hospital of CITIC‐XIANGYAChangShaChina,Clinical Research Center for Reproduction and Genetics in Hunan ProvinceChangShaChina,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning CommissionChangShaChina
| | - Yueqiu Tan
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical ScienceCentral South UniversityChangShaChina,Reproductive and Genetic Hospital of CITIC‐XIANGYAChangShaChina,Clinical Research Center for Reproduction and Genetics in Hunan ProvinceChangShaChina,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning CommissionChangShaChina
| | - Guangxiu Lu
- Reproductive and Genetic Hospital of CITIC‐XIANGYAChangShaChina,Clinical Research Center for Reproduction and Genetics in Hunan ProvinceChangShaChina,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning CommissionChangShaChina
| | - Wei Zheng
- Reproductive and Genetic Hospital of CITIC‐XIANGYAChangShaChina,Clinical Research Center for Reproduction and Genetics in Hunan ProvinceChangShaChina
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical ScienceCentral South UniversityChangShaChina,Reproductive and Genetic Hospital of CITIC‐XIANGYAChangShaChina,Clinical Research Center for Reproduction and Genetics in Hunan ProvinceChangShaChina,Laboratory of Reproductive and Stem Cell Engineering, National Health and Family Planning CommissionChangShaChina
| |
Collapse
|
18
|
Qu X, Han Y, Chen X, Lv Y, Zhang Y, Cao L, Zhang J, Jin Y. Inhibition of 26 S Proteasome Enhances AKAP3-mediated cAMP-PKA Signaling During Boar Sperm Capacitation. Anim Reprod Sci 2022; 247:107079. [DOI: 10.1016/j.anireprosci.2022.107079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 08/20/2022] [Accepted: 09/21/2022] [Indexed: 11/01/2022]
|
19
|
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
|
20
|
Wiesehöfer C, Wiesehöfer M, Dankert JT, Chung JJ, von Ostau NE, Singer BB, Wennemuth G. CatSper and its CaM-like Ca 2+ sensor EFCAB9 are necessary for the path chirality of sperm. FASEB J 2022; 36:e22288. [PMID: 35438819 PMCID: PMC9835897 DOI: 10.1096/fj.202101656rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 01/14/2023]
Abstract
Successful fertilization depends on sperm motility adaptation. Ejaculated and activated sperm beat symmetrically in high frequency, move linearly, and swim with clockwise chirality. After capacitation, sperm beat asymmetrically with lower amplitude and a high lateral head excursion. This motility change called hyperactivation requires CatSper activation and an increase in intracellular Ca2+ . However, whether CatSper-mediated Ca2+ influx participates in controlling the swim path chirality is unknown. In this study, we show that the clockwise path chirality is preserved in mouse sperm regardless of capacitation state but is lost in the sperm either lacking the entire CatSper channel or its Ca2+ sensor EFCAB9. Pharmacological inhibition of CatSper with either mibefradil or NNC 55-0396 leads to the same loss in swim path chirality. Exposure of sperm to the recombinant N-terminal part of the zona pellucida protein 2 randomizes chirality in capacitated cells, but not in non-capacitated ones. We conclude that Ca2+ sensitive regulation of CatSper activity orchestrates clockwise swim path chirality of sperm and any substantial change, such as the physiological stimulus of zona pellucida glycoproteins, results in a loss of chirality.
Collapse
Affiliation(s)
| | - Marc Wiesehöfer
- Department of Anatomy, University Duisburg-Essen, D-45147 Essen, Germany
| | | | - Jean-Ju Chung
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Nicola Edith von Ostau
- Department of Anatomy, University Duisburg-Essen, D-45147 Essen, Germany,Department of Urology, University Hospital Essen, D-45147 Essen, Germany
| | | | - Gunther Wennemuth
- Department of Anatomy, University Duisburg-Essen, D-45147 Essen, Germany,Correspondence to
| |
Collapse
|
21
|
Shi C, Zhang J, Yan Z, Gao L, Gao C, Wu W, Liu J, Cui Y. Epigenetic effect of putrescine supplementation during in vitro maturation of oocytes on offspring in mice. J Assist Reprod Genet 2022; 39:681-694. [PMID: 35254568 PMCID: PMC8995222 DOI: 10.1007/s10815-022-02448-6] [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: 12/08/2021] [Accepted: 02/25/2022] [Indexed: 01/12/2023] Open
Abstract
PURPOSE To investigate the epigenetic safety of putrescine supplementation during in vitro maturation (IVM) to offspring. METHODS Germinal vesicle oocytes retrieved from 12-week-old mice were randomly divided into two groups and cultured in IVM medium with or without 1 mmol/L putrescine for 16 h. Then, in vitro fertilization and embryo transplantation were conducted to produce the F1 offspring. The F1 mated with ordinary mice and bred the F2 offspring. The DNA methylation patterns in the brain and heart of F1 were investigated by reduced representation bisulfite sequencing. Imprinted gene expression levels of F1 oocytes were tested. The global methylation of F2 was examined by dot blot. RESULTS The weight, organ coefficient, and histology were normal in the F1 and F2 offspring from the putrescine-treated oocytes. An overall methylation level of 31.23 to 32.53% was observed for all CpG sites in the brain and heart of the two groups. The DNA methylation patterns of the brain and heart in F1 were not altered in general, with subtle differences. The expression levels of imprinted genes including H19, Snrpn, Peg3, Igf2, and Igf2r did not statistically change. The global 5mC level of F2 was consistent with the control group. CONCLUSION Putrescine supplementation during IVM did not directly affect the development, health, and reproduction, and did not affect the genome and global epigenetics of mouse offspring derived from those oocytes. The transient putrescine treatment for improving oocyte maturation shows its long-term safety of genome and epigenetics in the offspring of mice.
Collapse
Affiliation(s)
- Chennan Shi
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Jingyi Zhang
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Zhengjie Yan
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Li Gao
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Chao Gao
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Wei Wu
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Jiayin Liu
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Yugui Cui
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
22
|
Hwang JY, Wang H, Lu Y, Ikawa M, Chung JJ. C2cd6-encoded CatSperτ targets sperm calcium channel to Ca 2+ signaling domains in the flagellar membrane. Cell Rep 2022; 38:110226. [PMID: 34998468 PMCID: PMC8857959 DOI: 10.1016/j.celrep.2021.110226] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/29/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
In mammalian sperm cells, regulation of spatiotemporal Ca2+ signaling relies on the quadrilinear Ca2+ signaling nanodomains in the flagellar membrane. The sperm-specific, multi-subunit CatSper Ca2+ channel, which is crucial for sperm hyperactivated motility and male fertility, organizes the nanodomains. Here, we report CatSperτ, the C2cd6-encoded membrane-associating C2 domain protein, can independently migrate to the flagella and serve as a major targeting component of the CatSper channel complex. CatSperτ loss of function in mice demonstrates that it is essential for sperm hyperactivated motility and male fertility. CatSperτ targets the CatSper channel into the quadrilinear nanodomains in the flagella of developing spermatids, whereas it is dispensable for functional channel assembly. CatSperτ interacts with ciliary trafficking machinery in a C2-dependent manner. These findings provide insights into the CatSper channel trafficking to the Ca2+ signaling nanodomains and the shared molecular mechanisms of ciliary and flagellar membrane targeting.
Collapse
Affiliation(s)
- Jae Yeon Hwang
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Huafeng Wang
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yonggang Lu
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871, Japan
| | - Jean-Ju Chung
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Gynecology and Obstetrics, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
23
|
Yang F, Gracia Gervasi M, Orta G, Tourzani DA, De la Vega-Beltrán JL, Ruthel G, Darszon A, Visconti PE, Wang PJ. C2CD6 regulates targeting and organization of the CatSper calcium channel complex in sperm flagella. Development 2022; 149:dev199988. [PMID: 34919125 PMCID: PMC8774747 DOI: 10.1242/dev.199988] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 12/01/2021] [Indexed: 11/20/2022]
Abstract
The CatSper cation channel is essential for sperm capacitation and male fertility. The multi-subunit CatSper complexes form highly organized calcium signaling nanodomains on flagellar membranes. Here, we report identification of an uncharacterized protein, C2CD6, as a subunit of the mouse CatSper complex. C2CD6 contains a calcium-dependent, membrane-targeting C2 domain. C2CD6 associates with the CatSper calcium-selective, core-forming subunits. Deficiency of C2CD6 depletes the CatSper nanodomains from the flagellum and results in male sterility. C2CD6-deficient sperm are defective in hyperactivation and fail to fertilize oocytes both in vitro and in vivo. CatSper currents are present but at a significantly lower level in C2CD6-deficient sperm. Transient treatments with either Ca2+ ionophore, starvation, or a combination of both restore the fertilization capacity of C2CD6-deficient sperm. C2CD6 interacts with EFCAB9, a pH-dependent calcium sensor in the CatSper complex. We postulate that C2CD6 facilitates incorporation of the CatSper complex into the flagellar plasma membrane and may function as a calcium sensor. The identification of C2CD6 may enable the long-sought reconstitution of the CatSper ion channel complex in a heterologous system for male contraceptive development.
Collapse
Affiliation(s)
- Fang Yang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Maria Gracia Gervasi
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Gerardo Orta
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | - Darya A. Tourzani
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jose Luis De la Vega-Beltrán
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | - Gordon Ruthel
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | - Pablo E. Visconti
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - P. Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| |
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
|
Staicu FD, Martínez-Soto JC, Canovas S, Matás C. Nitric oxide-targeted protein phosphorylation during human sperm capacitation. Sci Rep 2021; 11:20979. [PMID: 34697378 PMCID: PMC8546126 DOI: 10.1038/s41598-021-00494-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 10/07/2021] [Indexed: 12/25/2022] Open
Abstract
Among many other molecules, nitric oxide insures the correct progress of sperm capacitation by mediating phosphorylation events. For a more comprehensive understanding of how this happens, we capacitated human spermatozoa from healthy men in the presence/absence of S-Nitrosoglutathione, a nitric oxide donor, two nitric oxide synthase inhibitors, NG-Nitro-l-arginine Methyl Ester Hydrochloride and Aminoguanidine Hemisulfate salt and, finally, with/without l-Arginine, the substrate for nitric oxide synthesis, and/or human follicular fluid. When analyzing the phosphorylation of protein kinase A substrates and tyrosine residues, we particularly observed how the inhibition of nitric oxide synthesis affects certain protein bands (~ 110, ~ 87, ~ 75 and ~ 62 kD) by lowering their phosphorylation degree, even when spermatozoa were incubated with l-Arginine and/or follicular fluid. Mass spectrometry analysis identified 29 proteins in these species, related to: spermatogenesis, binding to the zona pellucida, energy and metabolism, stress response, motility and structural organization, signaling and protein turnover. Significant changes in the phosphorylation degree of specific proteins could impair their biological activity and result in severe fertility-related phenotypes. These findings provide a deeper understanding of nitric oxide’s role in the capacitation process, and consequently, future studies in infertile patients should determine how nitric oxide mediates phosphorylation events in the species here described.
Collapse
Affiliation(s)
- Florentin-Daniel Staicu
- Department of Physiology, Veterinary Faculty, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Calle Campus Universitario, 11, 30100, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB), Murcia, Spain
| | | | - Sebastian Canovas
- Institute for Biomedical Research of Murcia (IMIB), Murcia, Spain.,Department of Physiology, Nursery Faculty, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain
| | - Carmen Matás
- Department of Physiology, Veterinary Faculty, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Calle Campus Universitario, 11, 30100, Murcia, Spain. .,Institute for Biomedical Research of Murcia (IMIB), Murcia, Spain.
| |
Collapse
|
26
|
Björkgren I, Chung DH, Mendoza S, Gabelev-Khasin L, Petersen NT, Modzelewski A, He L, Lishko PV. Alpha/Beta Hydrolase Domain-Containing Protein 2 Regulates the Rhythm of Follicular Maturation and Estrous Stages of the Female Reproductive Cycle. Front Cell Dev Biol 2021; 9:710864. [PMID: 34568325 PMCID: PMC8455887 DOI: 10.3389/fcell.2021.710864] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/09/2021] [Indexed: 12/26/2022] Open
Abstract
Mammalian female fertility is defined by a successful and strictly periodic ovarian cycle, which is under the control of gonadotropins and steroid hormones, particularly progesterone and estrogen. The latter two are produced by the ovaries that are engaged in controlled follicular growth, maturation, and release of the eggs, i.e., ovulation. The steroid hormones regulate ovarian cycles via genomic signaling, by altering gene transcription and protein synthesis. However, despite this well-studied mechanism, steroid hormones can also signal via direct, non-genomic action, by binding to their membrane receptors. Here we show, that the recently discovered membrane progesterone receptor α/β hydrolase domain-containing protein 2 (ABHD2) is highly expressed in mammalian ovaries where the protein plays a novel regulatory role in follicle maturation and the sexual cycle of females. Ablation of Abhd2 caused a dysregulation of the estrous cycle rhythm with females showing shortened luteal stages while remaining in the estrus stage for a longer time. Interestingly, the ovaries of Abhd2 knockout (KO) females resemble polycystic ovary morphology (PCOM) with a high number of atretic antral follicles that could be rescued with injection of gonadotropins. Such a procedure also allowed Abhd2 KO females to ovulate a significantly increased number of mature and fertile eggs in comparison with their wild-type littermates. These results suggest a novel regulatory role of ABHD2 as an important factor in non-genomic steroid regulation of the female reproductive cycle.
Collapse
Affiliation(s)
- Ida Björkgren
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Dong Hwa Chung
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Sarah Mendoza
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Liliya Gabelev-Khasin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Natalie T. Petersen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Andrew Modzelewski
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Lin He
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Polina V. Lishko
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
- The Center for Reproductive Longevity and Equality at the Buck Institute for Research on Aging, Novato, CA, United States
| |
Collapse
|
27
|
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
|
28
|
Luque GM, Xu X, Romarowski A, Gervasi MG, Orta G, De la Vega-Beltrán JL, Stival C, Gilio N, Dalotto-Moreno T, Krapf D, Visconti PE, Krapf D, Darszon A, Buffone MG. Cdc42 localized in the CatSper signaling complex regulates cAMP-dependent pathways in mouse sperm. FASEB J 2021; 35:e21723. [PMID: 34224609 DOI: 10.1096/fj.202002773rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 11/11/2022]
Abstract
Sperm acquire the ability to fertilize in a process called capacitation and undergo hyperactivation, a change in the motility pattern, which depends on Ca2+ transport by CatSper channels. CatSper is essential for fertilization and it is subjected to a complex regulation that is not fully understood. Here, we report that similar to CatSper, Cdc42 distribution in the principal piece is confined to four linear domains and this localization is disrupted in CatSper1-null sperm. Cdc42 inhibition impaired CatSper activity and other Ca2+ -dependent downstream events resulting in a severe compromise of the sperm fertilizing potential. We also demonstrate that Cdc42 is essential for CatSper function by modulating cAMP production by soluble adenylate cyclase (sAC), providing a new regulatory mechanism for the stimulation of CatSper by the cAMP-dependent pathway. These results reveal a broad mechanistic insight into the regulation of Ca2+ in mammalian sperm, a matter of critical importance in male infertility as well as in contraception.
Collapse
Affiliation(s)
- Guillermina M Luque
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Xinran Xu
- Department of Electrical and Computer Engineering, Colorado State University, Fort Collins, CO, USA
| | - Ana Romarowski
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina.,Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA, USA
| | - María G Gervasi
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA, USA
| | - Gerardo Orta
- Instituto de Biotecnología, UNAM, Cuernavaca, México
| | | | - Cintia Stival
- Instituto de Biología Molecular y Celular de Rosario (CONICET-UNR), Rosario, Santa Fe, Argentina
| | - Nicolás Gilio
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Tomás Dalotto-Moreno
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Dario Krapf
- Instituto de Biología Molecular y Celular de Rosario (CONICET-UNR), Rosario, Santa Fe, Argentina
| | - Pablo E Visconti
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA, USA
| | - Diego Krapf
- Department of Electrical and Computer Engineering, Colorado State University, Fort Collins, CO, USA
| | | | - Mariano G Buffone
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
29
|
Sánchez-Cárdenas C, Romarowski A, Orta G, De la Vega-Beltrán JL, Martín-Hidalgo D, Hernández-Cruz A, Visconti PE, Darszon A. Starvation induces an increase in intracellular calcium and potentiates the progesterone-induced mouse sperm acrosome reaction. FASEB J 2021; 35:e21528. [PMID: 33742713 DOI: 10.1096/fj.202100122r] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/26/2022]
Abstract
We have recently reported two different methodologies that improve sperm functionality. The first method involved transient exposure to the Ca2+ ionophore A23187 , and the second required sperm incubation in the absence of energy nutrients (starvation). Both methods were associated with an initial loss of motility followed by a rescue step involving ionophore removal or addition of energy metabolites, respectively. In this work, we show that starvation is accompanied by an increase in intracellular Ca2+ ([Ca2+ ]i ). Additionally, the starved cells acquire a significantly enhanced capacity to undergo a progesterone-induced acrosome reaction. Electrophysiological measurements show that CatSper channel remains active in starvation conditions. However, the increase in [Ca2+ ]i was also observed in sperm from CatSper null mice. Upon starvation, addition of energy nutrients reversed the effects on [Ca2+ ]i and decreased the effect of progesterone on the acrosome reaction to control levels. These data indicate that both methods have common molecular features.
Collapse
Affiliation(s)
- Claudia Sánchez-Cárdenas
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, México
| | - Ana Romarowski
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Gerardo Orta
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, México
| | - José Luis De la Vega-Beltrán
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, México
| | - David Martín-Hidalgo
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA.,Research Group of Intracellular Signalling and Technology of Reproduction (Research Institute INBIO G+C), University of Extremadura, Cáceres, Spain
| | - Arturo Hernández-Cruz
- Departamento de Neurociencia Cognitiva and Laboratorio Nacional de Canalopatías, Instituto de Fisiología Celular, UNAM, Ciudad Universitaria, México, México
| | - Pablo E Visconti
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, México
| |
Collapse
|
30
|
Curci L, Carvajal G, Sulzyk V, Gonzalez SN, Cuasnicú PS. Pharmacological Inactivation of CatSper Blocks Sperm Fertilizing Ability Independently of the Capacitation Status of the Cells: Implications for Non-hormonal Contraception. Front Cell Dev Biol 2021; 9:686461. [PMID: 34295893 PMCID: PMC8290173 DOI: 10.3389/fcell.2021.686461] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/26/2021] [Indexed: 12/28/2022] Open
Abstract
Cation channel of sperm (CatSper), the main sperm-specific Ca2+ channel, plays a key role in mammalian fertilization, and it is essential for male fertility, becoming an attractive target for contraception. Based on this, in the present work, we investigated the effects of CatSper inactivation on in vitro and in vivo sperm fertilizing ability and the mechanisms underlying such effects. Exposure of cauda epididymal mouse sperm to different concentrations (1-20 μM) of the potent CatSper inhibitor HC-056456 (HC) during in vitro capacitation showed no effects on sperm viability but significantly affected Ca2+ entry into the cells, progressive motility, protein tyrosine phosphorylation, induced acrosome reaction, and hyperactivation, as well as the sperm's ability to in vitro fertilize cumulus oocyte complexes and zona-free eggs. Whereas the presence of HC during gamete coincubation did not affect in vitro fertilization, exposure of either non-capacitating or already capacitated sperm to HC prior to gamete coincubation severely reduced fertilization, indicating that sperm function is affected by HC when the cells are incubated with the drug before sperm-egg interaction. Of note, insemination of HC-treated sperm into the uterus significantly or completely reduced the percentage of oviductal fertilized eggs showing, for the first time, the effects of a CatSper inhibitor on in vivo fertilization. These observations, together with the finding that HC affects sperm fertilizing ability independently of the sperm capacitation status, provide further insights on how CatSper regulates sperm function and represent a solid proof of concept for developing a male/female non-hormonal contraceptive based on the pharmacological blockage of CatSper activity.
Collapse
Affiliation(s)
- Ludmila Curci
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Guillermo Carvajal
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Valeria Sulzyk
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Soledad Natalia Gonzalez
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Patricia S Cuasnicú
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
31
|
Bianchi E, Stermer A, Nolan T, Li H, Hall S, Boekelheide K, Sigman M, Hwang K. Highly conserved sperm function-related transcripts across three species: human, rat and mouse. Reprod Toxicol 2021; 104:44-51. [PMID: 34174366 DOI: 10.1016/j.reprotox.2021.06.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 01/24/2023]
Abstract
Assessing male reproductive toxicity of environmental and therapeutic agents relies on the histopathology of the testis and epididymis in a pre-clinical setting. Animal histopathology poorly correlates with human sperm parameters, and none of these current methods are strong indicators of sperm health or reproductive potential. Therefore, there is an urgent need to identify a translatable, non-invasive and reliable approach to monitor environmental and therapeutic agents' effects on male reproductive health. mRNA sequences were analyzed in mouse, rat and human sperm samples to identify sperm transcriptomic similarities across species that could be used as biomarkers to predict male reproductive toxicity in animal models. Semen specimens were collected from men aged 18 to 55 years with proven fertility. Rat and mouse semen specimens were collected via needle punctures of the cauda epididymides. Sperm RNAs were extracted using an optimized sperm RNA isolation protocol and subjected to polyA-purified mRNA-sequencing. Bioinformatics analyses, including differential abundance and gene set enrichment analysis, were used to investigate the biological and molecular functions of all shared and differentially abundant transcripts across species. Transcriptome profiling identified 6,684 similarly expressed transcripts within the three species of which 1,579 transcripts were found to be involved in spermatogenic functions. Our findings have shown that sperm transcriptome is highly species dependent, however, there are some key similarities among transcripts that are required for fertility. Based on these similarities, sperm mRNA biomarker may be developed to monitor male reproductive toxicity where rodent models would make suitable laboratory substitutes for human.
Collapse
Affiliation(s)
- Enrica Bianchi
- Division of Urology, Rhode Island Hospital, Providence, RI, USA; Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Angela Stermer
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Timothy Nolan
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Hui Li
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Susan Hall
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Mark Sigman
- Division of Urology, Rhode Island Hospital, Providence, RI, USA; Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Kathleen Hwang
- Department of Urology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
32
|
Hwang JY, Maziarz J, Wagner GP, Chung JJ. Molecular Evolution of CatSper in Mammals and Function of Sperm Hyperactivation in Gray Short-Tailed Opossum. Cells 2021; 10:cells10051047. [PMID: 33946695 PMCID: PMC8147001 DOI: 10.3390/cells10051047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 12/25/2022] Open
Abstract
Males have evolved species-specifical sperm morphology and swimming patterns to adapt to different fertilization environments. In eutherians, only a small fraction of the sperm overcome the diverse obstacles in the female reproductive tract and successfully migrate to the fertilizing site. Sperm arriving at the fertilizing site show hyperactivated motility, a unique motility pattern displaying asymmetric beating of sperm flagella with increased amplitude. This motility change is triggered by Ca2+ influx through the sperm-specific ion channel, CatSper. However, the current understanding of the CatSper function and its molecular regulation is limited in eutherians. Here, we report molecular evolution and conservation of the CatSper channel in the genome throughout eutherians and marsupials. Sequence analyses reveal that CatSper proteins are slowly evolved in marsupials. Using an American marsupial, gray short-tailed opossum (Monodelphis domestica), we demonstrate the expression of CatSper in testes and its function in hyperactivation and unpairing of sperm. We demonstrate that a conserved IQ-like motif in CatSperζ is required for CatSperζ interaction with the pH-tuned Ca2+ sensor, EFCAB9, for regulating CatSper activity. Recombinant opossum EFCAB9 can interact with mouse CatSperζ despite high sequence divergence of CatSperζ among CatSper subunits in therians. Our finding suggests that molecular characteristics and functions of CatSper are evolutionarily conserved in gray short-tailed opossum, unraveling the significance of sperm hyperactivation and fertilization in marsupials for the first time.
Collapse
Affiliation(s)
- Jae Yeon Hwang
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
- Correspondence: (J.Y.H.); (J.-J.C.)
| | - Jamie Maziarz
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520, USA; (J.M.); (G.P.W.)
- Yale Systems Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Günter P. Wagner
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520, USA; (J.M.); (G.P.W.)
- Yale Systems Biology Institute, Yale University, West Haven, CT 06516, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jean-Ju Chung
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
- Correspondence: (J.Y.H.); (J.-J.C.)
| |
Collapse
|
33
|
Zhang X, Yu K, Ma L, Qian Z, Tian X, Miao Y, Niu Y, Xu X, Guo S, Yang Y, Wang Z, Xue X, Gu C, Fang W, Sun J, Yu Y, Wang J. Endogenous glutamate determines ferroptosis sensitivity via ADCY10-dependent YAP suppression in lung adenocarcinoma. Am J Cancer Res 2021; 11:5650-5674. [PMID: 33897873 PMCID: PMC8058707 DOI: 10.7150/thno.55482] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/03/2021] [Indexed: 12/22/2022] Open
Abstract
Rationale: Ferroptosis, a newly identified form of regulated cell death, can be induced following the inhibition of cystine-glutamate antiporter system XC- because of the impaired uptake of cystine. However, the outcome following the accumulation of endogenous glutamate in lung adenocarcinoma (LUAD) has not yet been determined. Yes-associated protein (YAP) is sustained by the hexosamine biosynthesis pathway (HBP)-dependent O-linked beta-N-acetylglucosaminylation (O-GlcNAcylation), and glutamine-fructose-6-phosphate transaminase (GFPT1), the rate-limiting enzyme of the HBP, can be phosphorylated and inhibited by adenylyl cyclase (ADCY)-mediated activation of protein kinase A (PKA). However, whether accumulated endogenous glutamate determines ferroptosis sensitivity by influencing the ADCY/PKA/HBP/YAP axis in LUAD cells is not understood. Methods: Cell viability, cell death and the generation of lipid reactive oxygen species (ROS) and malondialdehyde (MDA) were measured to evaluate the responses to the induction of ferroptosis following the inhibition of system XC-. Tandem mass tags (TMTs) were employed to explore potential factors critical for the ferroptosis sensitivity of LUAD cells. Immunoblotting (IB) and quantitative RT-PCR (qPCR) were used to analyze protein and mRNA expression. Co-immunoprecipitation (co-IP) assays were performed to identify protein-protein interactions and posttranslational modifications. Metabolite levels were measured using the appropriate kits. Transcriptional regulation was evaluated using a luciferase reporter assay, chromatin immunoprecipitation (ChIP), and electrophoretic mobility shift assay (EMSA). Drug administration and limiting dilution cell transplantation were performed with cell-derived xenograft (CDX) and patient-derived xenograft (PDX) mouse models. The associations among clinical outcome, drug efficacy and ADCY10 expression were determined based on data from patients who underwent curative surgery and evaluated with patient-derived primary LUAD cells and tissues. Results: The accumulation of endogenous glutamate following system XC- inhibition has been shown to determine ferroptosis sensitivity by suppressing YAP in LUAD cells. YAP O-GlcNAcylation and expression cannot be sustained in LUAD cells upon impairment of GFPT1. Thus, Hippo pathway-like phosphorylation and ubiquitination of YAP are enhanced. ADCY10 acts as a key downstream target and diversifies the effects of glutamate on the PKA-dependent suppression of GFPT1. We also discovered that the protumorigenic and proferroptotic effects of ADCY10 are mediated separately. Advanced-stage LUADs with high ADCY10 expression are sensitive to ferroptosis. Moreover, LUAD cells with acquired therapy resistance are also prone to higher ADCY10 expression and are more likely to respond to ferroptosis. Finally, a varying degree of secondary labile iron increase is caused by the failure to sustain YAP-stimulated transcriptional compensation for ferritin at later stages further explains why ferroptosis sensitivity varies among LUAD cells. Conclusions: Endogenous glutamate is critical for ferroptosis sensitivity following the inhibition of system XC- in LUAD cells, and ferroptosis-based treatment is a good choice for LUAD patients with later-stage and/or therapy-resistant tumors.
Collapse
|
34
|
Rodríguez-Tobón E, Fierro R, González-Márquez H, García-Vázquez FA, Arenas-Ríos E. Boar sperm incubation with reduced glutathione (GSH) differentially modulates protein tyrosine phosphorylation patterns and reorganization of calcium in sperm, in vitro fertilization, and embryo development depending on concentrations. Res Vet Sci 2020; 135:386-396. [PMID: 33153763 DOI: 10.1016/j.rvsc.2020.10.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022]
Abstract
The sperm in the female's reproductive tract undergo changes to fertilize the oocyte (sperm capacitation). These changes are regulated by redox system. However, some assisted reproductive technologies require sperm capacitation under in vitro conditions, though this increases the generation of ROS. Therefore, the aim of this study was to evaluate the effect of GSH as an antioxidant agent during the capacitation of boar sperm [evaluated by calcium compartmentalization, tyrosine phosphorylation (Tyr-P), motility, viability, and acrosomal integrity], in vitro fertilization (evaluated by penetration, monospermy, and efficiency %), and later embryo development (evaluated by cleavage and blastocyst rates, total number of cells per blastocyst and blastocyst diameter). Four experimental groups with different GSH concentrations (0-control, 0.5, 1, and 5 mM) were formed. When 1-GSH was added to the medium, the percentage of capacitated sperm increased after 4 h of incubation; the localization of Tyr-P was modified at 1 h and 4 h of incubation depending on the GSH concentration. Percentages of total and progressive sperm motility also increased at 4 h of incubation, but only in the 5-GSH group compared to control. Viability, acrosomal integrity, and general Tyr-P (Western blot) not differ among the experimental groups. The addition of GSH during gamete interaction increased penetration, monospermy, and efficiency rates in the 1-GSH group compared to the others. However, the effect of GSH was not observed in cleavage and blastocyst rates compared to the control. In conclusion, adding GSH modulates sperm capacitation (by means of calcium compartmentalization and tyrosine phosphorilation pattern) depending on its concentration, and improves IVF output at 1-GSH during gamete interaction.
Collapse
Affiliation(s)
- Ernesto Rodríguez-Tobón
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, CDMX, Mexico
| | - Reyna Fierro
- Universidad Autónoma Metropolitana, Departamento de Ciencias de la Salud, Unidad Iztapalapa, CDMX, Mexico.
| | - Humberto González-Márquez
- Universidad Autónoma Metropolitana, Departamento de Ciencias de la Salud, Unidad Iztapalapa, CDMX, Mexico.
| | - Francisco A García-Vázquez
- Departamento de Fisiología, Facultad de Veterinaria, Campus Internacional de Excelencia para la educación superior e investigación "Campus Mare Nostrum", Universidad de Murcia, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain.
| | - Edith Arenas-Ríos
- Universidad Autónoma Metropolitana, Departamento de Biología de la Reproducción, Unidad Iztapalapa, CDMX, Mexico.
| |
Collapse
|
35
|
The Role of Zinc in Male Fertility. Int J Mol Sci 2020; 21:ijms21207796. [PMID: 33096823 PMCID: PMC7589359 DOI: 10.3390/ijms21207796] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 12/18/2022] Open
Abstract
Several studies proposed the importance of zinc ion in male fertility. Here, we describe the properties, roles and cellular mechanisms of action of Zn2+ in spermatozoa, focusing on its involvement in sperm motility, capacitation and acrosomal exocytosis, three functions that are crucial for successful fertilization. The impact of zinc supplementation on assisted fertilization techniques is also described. The impact of zinc on sperm motility has been investigated in many vertebrate and invertebrate species. It has been reported that Zn2+ in human seminal plasma decreases sperm motility and that Zn2+ removal enhances motility. Reduction in the intracellular concentration of Zn2+ during epididymal transit allows the development of progressive motility and the subsequent hyper activated motility during sperm capacitation. Extracellular Zn2+ affects intracellular signaling pathways through its interaction with the Zn2+ sensing receptor (ZnR), also named GPR39. This receptor was found in the sperm tail and the acrosome, suggesting the possible involvement of Zn2+ in sperm motility and acrosomal exocytosis. Our studies showed that Zn2+ stimulates bovine sperm acrosomal exocytosis, as well as human sperm hyper-activated motility, were both mediated by GPR39. Zn2+ binds and activates GPR39, which activates the trans-membrane-adenylyl-cyclase (tmAC) to catalyze cAMP production. The NHE (Na+/H+-exchanger) is activated by cAMP, leading in increased pHi and activation of the sperm-specific Ca2+ channel CatSper, resulting in an increase in [Ca2+]i, which, together with HCO3−, activates the soluble adenylyl-cyclase (sAC). The increase in [cAMP]i activates protein kinase A (PKA), followed by activation of the Src-epidermal growth factor receptor-Pphospholipase C (Src-EGFR-PLC) cascade, resulting in inositol-triphosphate (IP3) production, which mobilizes Ca2+ from the acrosome, causing a further increase in [Ca2+]i and the development of hyper-activated motility. PKA also activates phospholipase D1 (PLD1), leading to F-actin formation during capacitation. Prior to the acrosomal exocytosis, PLC induces phosphadidylinositol-4,5-bisphosphate (PIP2) hydrolysis, leading to the release of the actin-severing protein gelsolin to the cytosol, which is activated by Ca2+, resulting in F-actin breakdown and the occurrence of acrosomal exocytosis.
Collapse
|
36
|
Ded L, Hwang JY, Miki K, Shi HF, Chung JJ. 3D in situ imaging of the female reproductive tract reveals molecular signatures of fertilizing spermatozoa in mice. eLife 2020; 9:62043. [PMID: 33078708 PMCID: PMC7707823 DOI: 10.7554/elife.62043] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022] Open
Abstract
Out of millions of ejaculated sperm, a few reach the fertilization site in mammals. Flagellar Ca2+ signaling nanodomains, organized by multi-subunit CatSper calcium channel complexes, are pivotal for sperm migration in the female tract, implicating CatSper-dependent mechanisms in sperm selection. Here using biochemical and pharmacological studies, we demonstrate that CatSper1 is an O-linked glycosylated protein, undergoing capacitation-induced processing dependent on Ca2+ and phosphorylation cascades. CatSper1 processing correlates with protein tyrosine phosphorylation (pY) development in sperm cells capacitated in vitro and in vivo. Using 3D in situ molecular imaging and ANN-based automatic detection of sperm distributed along the cleared female tract, we demonstrate that spermatozoa past the utero-tubal junction possess the intact CatSper1 signals. Together, we reveal that fertilizing mouse spermatozoa in situ are characterized by intact CatSper channel, lack of pY, and reacted acrosomes. These findings provide molecular insight into sperm selection for successful fertilization in the female reproductive tract.
Collapse
Affiliation(s)
- Lukas Ded
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, United States.,Laboratory of Reproductive Biology, Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Jae Yeon Hwang
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, United States
| | - Kiyoshi Miki
- Boston Children's Hospital, Boston, United States
| | - Huanan F Shi
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, United States
| | - Jean-Ju Chung
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, United States.,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, United States
| |
Collapse
|
37
|
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
|
38
|
Hlisníková H, Petrovičová I, Kolena B, Šidlovská M, Sirotkin A. Effects and Mechanisms of Phthalates' Action on Reproductive Processes and Reproductive Health: A Literature Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E6811. [PMID: 32961939 PMCID: PMC7559247 DOI: 10.3390/ijerph17186811] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022]
Abstract
The production of plastic products, which requires phthalate plasticizers, has resulted in the problems for human health, especially that of reproductive health. Phthalate exposure can induce reproductive disorders at various regulatory levels. The aim of this review was to compile the evidence concerning the association between phthalates and reproductive diseases, phthalates-induced reproductive disorders, and their possible endocrine and intracellular mechanisms. Phthalates may induce alterations in puberty, the development of testicular dysgenesis syndrome, cancer, and fertility disorders in both males and females. At the hormonal level, phthalates can modify the release of hypothalamic, pituitary, and peripheral hormones. At the intracellular level, phthalates can interfere with nuclear receptors, membrane receptors, intracellular signaling pathways, and modulate gene expression associated with reproduction. To understand and to treat the adverse effects of phthalates on human health, it is essential to expand the current knowledge concerning their mechanism of action in the organism.
Collapse
Affiliation(s)
- Henrieta Hlisníková
- Department of Zoology and Anthropology, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia; (I.P.); (B.K.); (M.Š.); (A.S.)
| | | | | | | | | |
Collapse
|
39
|
Hidalgo DM, Romarowski A, Gervasi MG, Navarrete F, Balbach M, Salicioni AM, Levin LR, Buck J, Visconti PE. Capacitation increases glucose consumption in murine sperm. Mol Reprod Dev 2020; 87:1037-1047. [PMID: 32914502 DOI: 10.1002/mrd.23421] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 08/28/2020] [Indexed: 12/15/2022]
Abstract
Mammalian sperm acquire fertilization capacity in the female reproductive tract in a process known as capacitation. During capacitation, sperm change their motility pattern (i.e., hyperactivation) and become competent to undergo the acrosome reaction. We have recently shown that, in the mouse, sperm capacitation is associated with increased uptake of fluorescently labeled deoxyglucose and with extracellular acidification suggesting enhanced glycolysis. Consistently, in the present work we showed that glucose consumption is enhanced in media that support mouse sperm capacitation suggesting upregulation of glucose metabolic pathways. The increase in glucose consumption was modulated by bicarbonate and blocked by protein kinase A and soluble adenylyl cyclase inhibitors. Moreover, permeable cyclic adenosine monophosphate (cAMP) agonists increase glucose consumption in sperm incubated in conditions that do not support capacitation. Also, the increase in glucose consumption was reduced when sperm were incubated in low calcium conditions. Interestingly, this reduction was not overcome with cAMP agonists. Despite these findings, glucose consumption of sperm from Catsper1 knockout mice was similar to the one from wild type suggesting that other sources of calcium are also relevant. Altogether, these results suggest that cAMP and calcium pathways are involved in the regulation of glycolytic energy pathways during murine sperm capacitation.
Collapse
Affiliation(s)
- David M Hidalgo
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA.,Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, Caceres, Spain
| | - Ana Romarowski
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA
| | - María G Gervasi
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA
| | - Felipe Navarrete
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA
| | - Melanie Balbach
- Department of Pharmacology, Weill Cornell New York, New York City, New York, USA
| | - Ana M Salicioni
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA
| | - Lonny R Levin
- Department of Pharmacology, Weill Cornell New York, New York City, New York, USA
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell New York, New York City, New York, USA
| | - Pablo E Visconti
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA
| |
Collapse
|
40
|
Luongo C, González-Brusi L, Cots-Rodríguez P, Izquierdo-Rico MJ, Avilés M, García-Vázquez FA. Sperm Proteome after Interaction with Reproductive Fluids in Porcine: From the Ejaculation to the Fertilization Site. Int J Mol Sci 2020; 21:ijms21176060. [PMID: 32842715 PMCID: PMC7570189 DOI: 10.3390/ijms21176060] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023] Open
Abstract
Ejaculated sperm are exposed to different environments before encountering the oocyte. However, how the sperm proteome changes during this transit remains unsolved. This study aimed to identify proteomic changes in boar sperm after incubation with male (seminal plasma, SP) and/or female (uterine fluid, UF; and oviductal fluid, OF) reproductive fluids. The following experimental groups were analyzed: (1) SP: sperm + 20% SP; (2) UF: sperm + 20% UF; (3) OF: sperm + 20% OF; (4) SP + UF: sperm + 20% SP + 20% UF; and (5) SP+OF: sperm + 20% SP + 20% OF. The proteome analysis, performed by HPLC-MS/MS, allowed the identification of 265 proteins. A total of 69 proteins were detected in the UF, SP, and SP + UF groups, and 102 proteins in the OF, SP, and SP + OF groups. Our results showed a higher number of proteins when sperm were incubated with only one fluid than when they were co-incubated with two fluids. Additionally, the number of sperm-interacting proteins from the UF group was lower than the OF group. In conclusion, the interaction of sperm with reproductive fluids alters its proteome. The description of sperm-interacting proteins in porcine species after co-incubation with male and/or female reproductive fluids may be useful to understand sperm transport, selection, capacitation, or fertilization phenomena.
Collapse
Affiliation(s)
- Chiara Luongo
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain;
| | - Leopoldo González-Brusi
- Department of Cell Biology and Histology, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain; (L.G.-B.); (P.C.-R.); (M.J.I.-R.)
| | - Paula Cots-Rodríguez
- Department of Cell Biology and Histology, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain; (L.G.-B.); (P.C.-R.); (M.J.I.-R.)
| | - Mª José Izquierdo-Rico
- Department of Cell Biology and Histology, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain; (L.G.-B.); (P.C.-R.); (M.J.I.-R.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
| | - Manuel Avilés
- Department of Cell Biology and Histology, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain; (L.G.-B.); (P.C.-R.); (M.J.I.-R.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
- Correspondence: (M.A.); (F.A.G.-V.)
| | - Francisco Alberto García-Vázquez
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain;
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
- Correspondence: (M.A.); (F.A.G.-V.)
| |
Collapse
|
41
|
Zhang J, Liu H, Yang Q, Li P, Wen Y, Han X, Li B, Jiang H, Li X. Genomic Sequencing Reveals the Diversity of Seminal Bacteria and Relationships to Reproductive Potential in Boar Sperm. Front Microbiol 2020; 11:1873. [PMID: 32903829 PMCID: PMC7438901 DOI: 10.3389/fmicb.2020.01873] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/16/2020] [Indexed: 02/01/2023] Open
Abstract
A number of emerging studies suggest that pathogenic microorganisms in semen may cause a decline in the reproductive potential of spermatozoa, and the bacterial diversity and profile of ejaculated boar semen in different seasons are currently unknown. To explore the bacterial composition and changes in ejaculated boar semen from winter and summer, and the underlying mechanism of decline in sperm quality and fertility capacity in summer, 120 ejaculated semen samples were examined for bacterial communities using genomic sequencing technology, and the associations between microbial composition and sperm reproductive potential were investigated. The results showed that Proteobacteria (57.53%), Firmicutes (31.17%), Bacteroidetes (4.24%), and Actinobacteria (3.41%) are the dominant phyla in the ejaculated semen, and the dominant genera were Pseudomonas (34.41%) and Lactobacillus (19.93%), which belong to the phyla of Proteobacteria and Firmicutes, respectively. Interestingly, the higher diversity of bacteria in ejaculated semen of winter differs from that of summer semen, potentially due to seasonal changes related to changes in semen quality and sperm fertilizing capacity. Furthermore, the highly abundant Lactobacillus in winter samples were positively associated with sperm quality and reproductive performance obtained from sows inseminated with such semen samples, while in contrast, the highly abundant Pseudomonas in summer samples was negatively associated with sperm quality and reproductive potential. Additionally, our results strongly indicated that Lactobacillus is not only a potential probiotic for semen quality and fertility potential but also beneficial for restraining the negative influence of Pseudomonas. Overall, our findings significantly contribute to the current understanding of the phenotypes and etiology of male "summer infertility," and may represent a frontier in male reproductive disorders and possible early prevention against pathogenic bacteria.
Collapse
Affiliation(s)
- Jing Zhang
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Huan Liu
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Qiangzhen Yang
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Peifei Li
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Wen
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Xuejun Han
- Shanghai Engineering Research Center of Breeding Pig, Shanghai, China
| | - Bushe Li
- Shanghai Engineering Research Center of Breeding Pig, Shanghai, China
| | - Hongju Jiang
- Shanghai Engineering Research Center of Breeding Pig, Shanghai, China
| | - Xinhong Li
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
42
|
Li K, Sun P, Wang Y, Gao T, Zheng D, Liu A, Ni Y. Hsp90 interacts with Cdc37, is phosphorylated by PKA/PKC, and regulates Src phosphorylation in human sperm capacitation. Andrology 2020; 9:185-195. [PMID: 32656999 DOI: 10.1111/andr.12862] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/26/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Heat shock protein 90 (Hsp90) signaling pathways participate in protein phosphorylation during sperm capacitation. However, the underlying mechanism is largely unknown. OBJECTIVE The aim of this study was to explore the interaction between Hsp90 and its co-chaperone protein, cell division cycle protein Cdc37 (Cdc37), in human spermatozoa. MATERIALS AND METHODS We examined the effects of H-89 (a protein kinase A [PKA] inhibitor) and Go6983 (a protein kinase C [PKC] inhibitor) on the phosphorylation of serine, threonine, and tyrosine residues in Hsp90; the effect of 17-allylamino-17-demethoxygeldanamycin (17-AAG, Hsp90 inhibitor) on Y416-Src phosphorylation; and the effects of 17-AAG and geldanamycin on threonine phosphorylation during human sperm capacitation. RESULTS Hsp90 co-localized and interacted with Cdc37. During human sperm capacitation, Hsp90 phosphorylation at serine, threonine, and tyrosine residues was inhibited by H-89 and Go6983. In addition, phosphorylation of residue Y416 in the tyrosine kinase Src (its active site) was inhibited by 17-AAG, and the threonine phosphorylation levels of some proteins were decreased by 17-AAG and geldanamycin. DISCUSSION AND CONCLUSION Taken together, our data showed that the interaction of Hsp90 with Cdc37 regulates total protein threonine phosphorylation and Src phosphorylation via its serine, threonine, and tyrosine phosphorylation, which are controlled by PKA and PKC during human sperm capacitation. The results of this study help understand the mechanism underlying Hsp90 regulation of sperm function.
Collapse
Affiliation(s)
- Kun Li
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Peibei Sun
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Yayan Wang
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Tian Gao
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Dongwang Zheng
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Ajuan Liu
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Ya Ni
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
43
|
Yang Q, Wen Y, Wang L, Peng Z, Yeerken R, Zhen L, Li P, Li X. Ca 2+ ionophore A23187 inhibits ATP generation reducing mouse sperm motility and PKA-dependent phosphorylation. Tissue Cell 2020; 66:101381. [PMID: 32933704 DOI: 10.1016/j.tice.2020.101381] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023]
Abstract
Male infertility is a global problem in modern society of which capacitating defects are a major cause. Previous studies have demonstrated that Ca2+ ionophore A23187 can make mouse sperm capable of fertilizing in vitro, which may aid in clinical treatment of capacitating defects. However, the detailed role and mechanism of Ca2+ in the capacitating process are still unclear especially how A23187 quickly renders sperm immotile and inhibits cAMP/PKA-mediated phosphorylation. We report that A23187 induces a Ca2+ flux in the mitochondria enriched sperm tail and excess Ca2+ inhibits key metabolic enzymes involved in acetyl-CoA biosynthesis, TCA cycle and electron transport chain pathways resulting in reduced ATP and overall energy production, however this flux does not destroy the structure of the sperm tail. Due to the decrease in ATP production, which is the main phosphate group donator and the power of sperm, the sperm is rendered immobile and PKA-mediated phosphorylation is inhibited. Our study proposed a possible mechanism through which A23187 reduces sperm motility and PKA-mediated phosphorylation from ATP generation, thus providing basic data for exploring the functional roles of Ca2+ in sperm in the future.
Collapse
Affiliation(s)
- Qiangzhen Yang
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yi Wen
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lirui Wang
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zijun Peng
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ranna Yeerken
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Linqing Zhen
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Peifei Li
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xinhong Li
- Shanghai Key Lab of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
44
|
Qu W, Yuan S, Quan C, Huang Q, Zhou Q, Yap Y, Shi L, Zhang D, Guest T, Li W, Yee SP, Zhang L, Cazin C, Hess RA, Ray PF, Kherraf ZE, Zhang Z. The essential role of intraflagellar transport protein IFT81 in male mice spermiogenesis and fertility. Am J Physiol Cell Physiol 2020; 318:C1092-C1106. [PMID: 32233951 DOI: 10.1152/ajpcell.00450.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intraflagellar transport (IFT) is an evolutionarily conserved mechanism that is indispensable for the formation and maintenance of cilia and flagella; however, the implications and functions of IFT81 remain unknown. In this study, we disrupted IFT81 expression in male germ cells starting from the spermatocyte stage. As a result, homozygous mutant males were completely infertile and displayed abnormal sperm parameters. In addition to oligozoospermia, spermatozoa presented dysmorphic and nonfunctional flagella. Histological examination of testes from homozygous mutant mice revealed abnormal spermiogenesis associated with sloughing of germ cells and the presence of numerous multinucleated giant germ cells (symblasts) in the lumen of seminiferous tubules and epididymis. Moreover, only few elongated spermatids and spermatozoa were seen in analyzed cross sections. Transmission electron microscopy showed a complete disorganization of the axoneme and para-axonemal structures such as the mitochondrial sheath, fibrous sheath, and outer dense fibers. In addition, numerous vesicles that contain unassembled microtubules were observed within developing spermatids. Acrosome structure analysis showed normal appearance, thus excluding a crucial role of IFT81 in acrosome biogenesis. These observations showed that IFT81 is an important member of the IFT process during spermatogenesis and that its absence is associated with abnormal flagellum formation leading to male infertility. The expression levels of several IFT components in testes, including IFT20, IFT25, IFT27, IFT57, IFT74, and IFT88, but not IFT140, were significantly reduced in homozygous mutant mice. Overall, our study demonstrates that IFT81 plays an essential role during spermatogenesis by modulating the assembly and elongation of the sperm flagella.
Collapse
Affiliation(s)
- Wei Qu
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Shuo Yuan
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Chao Quan
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Qian Huang
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Qi Zhou
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Yitian Yap
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Lin Shi
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - David Zhang
- College of William & Mary, Williamsburg, Virginia
| | - Tamia Guest
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Wei Li
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Siu-Pok Yee
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Ling Zhang
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Caroline Cazin
- Team Genetic, Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale U1209, Centre National de la Recherche Scientifique Unité Mixte de Recherche 5309, Grenoble, France.,Centre Hospitalier Universitaire de Grenoble Alpes, Grenoble, France
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, Illinois
| | - Pierre F Ray
- Team Genetic, Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale U1209, Centre National de la Recherche Scientifique Unité Mixte de Recherche 5309, Grenoble, France.,Centre Hospitalier Universitaire de Grenoble Alpes, Grenoble, France
| | - Zine-Eddine Kherraf
- Team Genetic, Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale U1209, Centre National de la Recherche Scientifique Unité Mixte de Recherche 5309, Grenoble, France.,Centre Hospitalier Universitaire de Grenoble Alpes, Grenoble, France
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, Detroit, Michigan.,Department of Obstetrics & Gynecology, Wayne State University, Detroit, Michigan
| |
Collapse
|
45
|
Paudel B, Gervasi MG, Porambo J, Caraballo DA, Tourzani DA, Mager J, Platt MD, Salicioni AM, Visconti PE. Sperm capacitation is associated with phosphorylation of the testis-specific radial spoke protein Rsph6a†. Biol Reprod 2020; 100:440-454. [PMID: 30239614 DOI: 10.1093/biolre/ioy202] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/03/2018] [Accepted: 09/13/2018] [Indexed: 12/31/2022] Open
Abstract
Mammalian sperm undergo a series of biochemical and physiological changes collectively known as capacitation in order to acquire the ability to fertilize. Although the increase in phosphorylation associated with mouse sperm capacitation is well established, the identity of the proteins involved in this signaling cascade remains largely unknown. Tandem mass spectrometry (MS/MS) has been used to identify the exact sites of phosphorylation and to compare the relative extent of phosphorylation at these sites. In the present work, we find that a novel site of phosphorylation on a peptide derived from the radial spoke protein Rsph6a is more phosphorylated in capacitated mouse sperm. The Rsph6a gene has six exons, five of which are conserved during evolution in flagellated cells. The exon containing the capacitation-induced phosphorylation site was found exclusively in eutherian mammals. Transcript analyses revealed at least two different testis-specific splicing variants for Rsph6a.Rsph6a mRNA expression was restricted to spermatocytes. Using antibodies generated against the Rsph6a N-terminal domain, western blotting and immunofluorescence analyses indicated that the protein remains in mature sperm and localizes to the sperm flagellum. Consistent with its role in the axoneme, solubility analyses revealed that Rsph6 is attached to cytoskeletal structures. Based on previous studies in Chlamydomonas reinhardtii, we predict that Rsph6 participates in the interaction between the central pair of microtubules and the surrounding pairs. The findings that Rsph6a is more phosphorylated during capacitation and is predicted to function in axonemal localization make Rsph6a a candidate protein mediating signaling processes in the sperm flagellum.
Collapse
Affiliation(s)
- Bidur Paudel
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA
| | - María Gracia Gervasi
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA
| | - James Porambo
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Diego A Caraballo
- IFIBYNE-CONICET, Laboratorio de Fisiología y Biología Molecular, Departamento de Fisiología, Biología Molecular y Celular, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Darya A Tourzani
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA
| | - Jesse Mager
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA
| | - Mark D Platt
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Ana María Salicioni
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA
| | - Pablo E Visconti
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts, Amherst, Massachusetts, USA
| |
Collapse
|
46
|
Leclerc P, Goupil S, Rioux J, Lavoie‐Ouellet C, Clark M, Ruiz J, Saindon A. Study on the role of calmodulin in sperm function through the enrichment and identification of calmodulin‐binding proteins in bovine ejaculated spermatozoa. J Cell Physiol 2020; 235:5340-5352. [DOI: 10.1002/jcp.29421] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/19/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Pierre Leclerc
- Department of Obstetrics, Gynecology and ReproductionCenter for Research on Reproduction, Development and Intergenerational Health, Laval University Quebec Canada
- Reproduction, Mother and Youth Health AxisCHU de Quebec‐Université Laval research Center Quebec Canada
| | - Serge Goupil
- Department of Obstetrics, Gynecology and ReproductionCenter for Research on Reproduction, Development and Intergenerational Health, Laval University Quebec Canada
- Reproduction, Mother and Youth Health AxisCHU de Quebec‐Université Laval research Center Quebec Canada
| | - Jean‐François Rioux
- Department of Obstetrics, Gynecology and ReproductionCenter for Research on Reproduction, Development and Intergenerational Health, Laval University Quebec Canada
- Reproduction, Mother and Youth Health AxisCHU de Quebec‐Université Laval research Center Quebec Canada
| | - Camille Lavoie‐Ouellet
- Department of Obstetrics, Gynecology and ReproductionCenter for Research on Reproduction, Development and Intergenerational Health, Laval University Quebec Canada
- Reproduction, Mother and Youth Health AxisCHU de Quebec‐Université Laval research Center Quebec Canada
| | - Marie‐Ève Clark
- Department of Obstetrics, Gynecology and ReproductionCenter for Research on Reproduction, Development and Intergenerational Health, Laval University Quebec Canada
- Reproduction, Mother and Youth Health AxisCHU de Quebec‐Université Laval research Center Quebec Canada
| | - Juliana Ruiz
- Department of Obstetrics, Gynecology and ReproductionCenter for Research on Reproduction, Development and Intergenerational Health, Laval University Quebec Canada
- Reproduction, Mother and Youth Health AxisCHU de Quebec‐Université Laval research Center Quebec Canada
| | - Andrée‐Anne Saindon
- Department of Obstetrics, Gynecology and ReproductionCenter for Research on Reproduction, Development and Intergenerational Health, Laval University Quebec Canada
- Reproduction, Mother and Youth Health AxisCHU de Quebec‐Université Laval research Center Quebec Canada
| |
Collapse
|
47
|
Mirnamniha M, Faroughi F, Tahmasbpour E, Ebrahimi P, Beigi Harchegani A. An overview on role of some trace elements in human reproductive health, sperm function and fertilization process. REVIEWS ON ENVIRONMENTAL HEALTH 2019; 34:339-348. [PMID: 31203261 DOI: 10.1515/reveh-2019-0008] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/28/2019] [Indexed: 05/28/2023]
Abstract
Human semen contains several trace elements such as calcium (Ca), copper (Cu), manganese (Mn), magnesium (Mg), zinc (Zn) and selenium (Se) which are necessary for reproductive health, normal spermatogenesis, sperm maturation, motility and capacitation, as well as normal sperm function. In this review, the potential role of these trace elements in male reproductive health, normal function of spermatozoa and fertility potency were considered. We selected and reviewed articles that considered crucial roles of trace elements in human sperm function and fertility. Ca is essential for sperm motility and its hyperactivation, sperm capacitation and acrosome reaction, as well as sperm chemotaxis. Sodium (Na) and potassium (K) are involved in sperm motility and capacitation. Mg is necessary for normal ejaculation, spermatogenesis and sperm motility. Zn is one of the most significant nutrients in human semen. Seminal deficiency of Zn can be associated with delayed testicular development, impaired spermatogenesis, deficiency of sex hormones, oxidative stress and inflammation, and apoptosis. Se is another significant element which has antioxidative properties and is essential for spermatogenesis and the maintenance of male fertility. Mn is a potent stimulator for sperm motility; however, increased level of seminal plasma Se can be toxic for sperm. Like Se, Cu has antioxidative properties and has a positive effect on sperm parameters. Decreased level of these trace elements can negatively affect human reproductive health, semen quality, sperm normal function and as the result, fertility potency in men. Measurement of these trace elements in men with idiopathic infertility is necessary.
Collapse
Affiliation(s)
- Mahdiyeh Mirnamniha
- Department of Medical Radiation Engineering, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Fereshteh Faroughi
- Department of Medical Radiation Engineering, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Eisa Tahmasbpour
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | - Pirooz Ebrahimi
- University Scientific Education and Research Network, Tehran, Iran
| | - Asghar Beigi Harchegani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19945-581, Tehran, Iran
| |
Collapse
|
48
|
Dey S, Brothag C, Vijayaraghavan S. Signaling Enzymes Required for Sperm Maturation and Fertilization in Mammals. Front Cell Dev Biol 2019; 7:341. [PMID: 31921853 PMCID: PMC6930163 DOI: 10.3389/fcell.2019.00341] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 12/03/2019] [Indexed: 12/18/2022] Open
Abstract
In mammals, motility and fertilizing ability of spermatozoa develop during their passage through the epididymis. After ejaculation, sperm undergo capacitation and hyperactivation in the female reproductive tract - a motility transition that is required for sperm penetration of the egg. Both epididymal initiation of sperm motility and hyperactivation are essential for male fertility. Motility initiation in the epididymis and sperm hyperactivation involve changes in metabolism, cAMP (cyclic adenosine mono-phosphate), calcium and pH acting through protein kinases and phosphatases. Despite this knowledge, we still do not understand, in biochemical terms, how sperm acquire motility in the epididymis and how motility is altered in the female reproductive tract. Recent data show that the sperm specific protein phosphatase PP1γ2, glycogen synthase kinase 3 (GSK3), and the calcium regulated phosphatase calcineurin (PP2B), are involved in epididymal sperm maturation. The protein phosphatase PP1γ2 is present only in testis and sperm in mammals. PP1γ2 has a isoform-specific requirement for normal function of mammalian sperm. Sperm PP1γ2 is regulated by three proteins - inhibitor 2, inhibitor 3 and SDS22. Changes in phosphorylation of these three inhibitors and their binding to PP1γ2 are involved in initiation and activation of sperm motility. The inhibitors are phosphorylated by protein kinases, one of which is GSK3. The isoform GSK3α is essential for epididymal sperm maturation and fertility. Calcium levels dramatically decrease during sperm maturation and initiation of motility suggesting that the calcium activated sperm phosphatase (PP2B) activity also decreases. Loss of PP2B results in male infertility due to impaired sperm maturation in the epididymis. Thus the three signaling enzymes PP1γ2, GSK3, and PP2B along with the documented PKA (protein kinase A) have key roles in sperm maturation and hyperactivation. Significantly, all these four signaling enzymes are present as specific isoforms only in placental mammals, a testimony to their essential roles in the unique aspects of sperm function in mammals. These findings should lead to a better biochemical understanding of the basis of male infertility and should lead to novel approaches to a male contraception and managed reproduction.
Collapse
|
49
|
Li X, Wang L, Liu H, Fu J, Zhen L, Li Y, Zhang Y, Zhang Y. C 60 Fullerenes Suppress Reactive Oxygen Species Toxicity Damage in Boar Sperm. NANO-MICRO LETTERS 2019; 11:104. [PMID: 34138040 PMCID: PMC7770955 DOI: 10.1007/s40820-019-0334-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 10/29/2019] [Indexed: 05/22/2023]
Abstract
We report the carboxylated C60 improved the survival and quality of boar sperm during liquid storage at 4 °C and thus propose the use of carboxylated C60 as a novel antioxidant semen extender supplement. Our results demonstrated that the sperm treated with 2 μg mL-1 carboxylated C60 had higher motility than the control group (58.6% and 35.4%, respectively; P ˂ 0.05). Moreover, after incubation with carboxylated C60 for 10 days, acrosome integrity and mitochondrial activity of sperm increased by 18.1% and 34%, respectively, compared with that in the control group. Similarly, the antioxidation abilities and adenosine triphosphate levels in boar sperm treated with carboxylated C60 significantly increased (P ˂ 0.05) compared with those in the control group. The presence of carboxylated C60 in semen extender increases sperm motility probably by suppressing reactive oxygen species (ROS) toxicity damage. Interestingly, carboxylated C60 could protect boar sperm from oxidative stress and energy deficiency by inhibiting the ROS-induced protein dephosphorylation via the cAMP-PKA signaling pathway. In addition, the safety of carboxylated C60 as an alternative antioxidant was also comprehensively evaluated by assessing the mean litter size and number of live offspring in the carboxylated C60 treatment group. Our findings confirm carboxylated C60 as a novel antioxidant agent and suggest its use as a semen extender supplement for assisted reproductive technology in domestic animals.
Collapse
Affiliation(s)
- Xinhong Li
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China.
| | - Lirui Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Huan Liu
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Jieli Fu
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Linqing Zhen
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Yuhua Li
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Yaozhong Zhang
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, USA
| | - Yafei Zhang
- Key Laboratory of Thin Film and Microfabrication (Ministry of Education), Department of Micro/Nano Electronics, School of Electronics, Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
50
|
Navarrete FA, Aguila L, Martin-Hidalgo D, Tourzani DA, Luque GM, Ardestani G, Garcia-Vazquez FA, Levin LR, Buck J, Darszon A, Buffone MG, Mager J, Fissore RA, Salicioni AM, Gervasi MG, Visconti PE. Transient Sperm Starvation Improves the Outcome of Assisted Reproductive Technologies. Front Cell Dev Biol 2019; 7:262. [PMID: 31750304 PMCID: PMC6848031 DOI: 10.3389/fcell.2019.00262] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/17/2019] [Indexed: 12/27/2022] Open
Abstract
To become fertile, mammalian sperm must undergo a series of biochemical and physiological changes known as capacitation. These changes involve crosstalk between metabolic and signaling pathways and can be recapitulated in vitro. In this work, sperm were incubated in the absence of exogenous nutrients (starved) until they were no longer able to move. Once immotile, energy substrates were added back to the media and sperm motility was rescued. Following rescue, a significantly higher percentage of starved sperm attained hyperactivated motility and displayed increased ability to fertilize in vitro when compared with sperm persistently incubated in standard capacitation media. Remarkably, the effects of this treatment continue beyond fertilization as starved and rescued sperm promoted higher rates of embryo development, and once transferred to pseudo-pregnant females, blastocysts derived from treated sperm produced significantly more pups. In addition, the starvation and rescue protocol increased fertilization and embryo development rates in sperm from a severely sub-fertile mouse model, and when combined with temporal increase in Ca2+ ion levels, this methodology significantly improved fertilization and embryo development rates in sperm of sterile CatSper1 KO mice model. Intracytoplasmic sperm injection (ICSI) does not work in the agriculturally relevant bovine system. Here, we show that transient nutrient starvation of bovine sperm significantly enhanced ICSI success in this species. These data reveal that the conditions under which sperm are treated impact post-fertilization development and suggest that this “starvation and rescue method” can be used to improve assisted reproductive technologies (ARTs) in other mammalian species, including humans.
Collapse
Affiliation(s)
- Felipe A Navarrete
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts Amherst, Amherst, MA, United States
| | - Luis Aguila
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts Amherst, Amherst, MA, United States
| | - David Martin-Hidalgo
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts Amherst, Amherst, MA, United States.,Research Group of Intracellular Signaling and Technology of Reproduction, Institute of Biotechnology in Agriculture and Livestock (INBIO G + C), University of Extremadura, Cáceres, Spain
| | - Darya A Tourzani
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts Amherst, Amherst, MA, United States
| | - Guillermina M Luque
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Goli Ardestani
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts Amherst, Amherst, MA, United States
| | - Francisco A Garcia-Vazquez
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research, University of Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia, IMIB-Arrixaca, Murcia, Spain
| | - Lonny R Levin
- Department of Pharmacology, Weill Cornell Medical College, Cornell University, New York, NY, United States
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medical College, Cornell University, New York, NY, United States
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Mariano G Buffone
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Jesse Mager
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts Amherst, Amherst, MA, United States
| | - Rafael A Fissore
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts Amherst, Amherst, MA, United States
| | - Ana M Salicioni
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts Amherst, Amherst, MA, United States
| | - María G Gervasi
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts Amherst, Amherst, MA, United States
| | - Pablo E Visconti
- Department of Veterinary and Animal Sciences, Integrated Sciences Building, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|