1
|
Dlamini NH, Bridi A, da Silveira JC, Feugang JM. Unlocking Gamete Quality Through Extracellular Vesicles: Emerging Perspectives. BIOLOGY 2025; 14:198. [PMID: 40001966 PMCID: PMC11851576 DOI: 10.3390/biology14020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025]
Abstract
Extracellular vesicles (EVs) are gaining recognition for their essential role in enhancing gamete quality and improving outcomes in assisted reproductive technologies. These nanosized particles, released by cells, carry proteins, lipids, and RNAs, facilitating critical cell communication and offering the potential to enhance gamete maturation and improve fertilization rates. Most research on males has concentrated on seminal plasma, a complex fluid produced by the testes and accessory glands vital in modulating sperm fertility potential. The components of seminal plasma significantly affect sperm functionality, embryo survival, and placental development, making this a prominent area of interest in reproductive biology. The EVs within seminal plasma contribute to maintaining sperm membrane stability, enhancing motility, and promoting capacitation, which may influence the female reproductive tract following mating. In females, EVs have been identified in both the follicular and uterine environments, where effective embryo-maternal communication is crucial. The oviduct epithelium supports gamete transport and early embryonic development, with EVs found in oviductal fluid playing a key role in reproductive processes. These EVs support the embryo's growth in the nutrient-rich uterine environment. These important studies underscore the significant role of EVs in transporting essential molecular compounds to gametes and embryos, leading to an enhanced understanding and potential manipulation of reproductive processes. This review aims to summarize the current research on the benefits of EVs in gamete manipulation and embryo development, highlighting their promising implications for reproductive health.
Collapse
Affiliation(s)
- Notsile H. Dlamini
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39759, USA;
| | - Alessandra Bridi
- University of the West of Santa Catarina, Xanxerê 89820-000, SC, Brazil;
| | | | - Jean M. Feugang
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39759, USA;
| |
Collapse
|
2
|
Weigel Muñoz M, Cohen DJ, Da Ros VG, González SN, Rebagliati Cid A, Sulzyk V, Cuasnicu PS. Physiological and pathological aspects of epididymal sperm maturation. Mol Aspects Med 2024; 100:101321. [PMID: 39340983 DOI: 10.1016/j.mam.2024.101321] [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: 07/08/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
In mammals, sperm that leave the testes are nonfunctional and require a complex post-testicular maturation process to acquire their ability to recognize and fertilize the egg. The crucial maturation changes that provide sperm their fertilizing capability occur while passing through the epididymis. Due to the widespread use of assisted reproductive technologies to address male infertility, there has been a significant decrease in research focusing on the mechanisms underlying the maturation process over the past decades. Considering that up to 40% of male infertility is idiopathic and could be reflecting sperm maturation defects, the study of post-testicular sperm maturation will clearly contribute to a better understanding of the causes of male infertility and to the development of both new approaches to maturing sperm in vitro and safer male contraceptive methods. Based on this, the present review focuses on the physiopathology of the epididymis as well as on current approaches under investigation to improve research in sperm maturation and as potential therapeutic options for male infertility.
Collapse
Affiliation(s)
- Mariana Weigel Muñoz
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Débora J Cohen
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Vanina G Da Ros
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Soledad N González
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Abril Rebagliati Cid
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Valeria Sulzyk
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Patricia S Cuasnicu
- Instituto de Biología y Medicina Experimental (IBYME). Fundación IBYME. Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
3
|
Rahbar M, Asadpour R, Mazaheri Z. The effect of epididymosomes on the development of frozen-thawed mouse spermatogonial stem cells after culture in a decellularized testicular scaffold and transplantation into azoospermic mice. J Assist Reprod Genet 2024; 41:2079-2098. [PMID: 38839698 PMCID: PMC11339233 DOI: 10.1007/s10815-024-03157-y] [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: 01/11/2024] [Accepted: 05/24/2024] [Indexed: 06/07/2024] Open
Abstract
PURPOSE This study examined SSC proliferation on an epididymosome-enriched decellularized testicular matrix (DTM) hydrogel and spermatogenesis induction in azoospermic mice. METHODS Epididymosomes were extracted and characterized using SEM and western blotting. After cryopreservation, thawed SSCs were cultured in a hydrogel-based three-dimensional (3D) culture containing 10 ng/mL GDNF or 20 µg/mL epididymosomes. SSCs were assessed using the MTT assay, flow cytometry, and qRT-PCR after two weeks of culture. The isolated SSCs were microinjected into the efferent ducts of busulfan-treated mice. DiI-labeled SSCs were followed, and cell homing was assessed after two weeks. After 8 weeks, the testes were evaluated using morphometric studies and immunohistochemistry. RESULTS The expression of PLZF, TGF-β, and miR-10b did not increase statistically significantly in the 3D + GDNF and 3D + epididymosome groups compared to the 3D group. Among the groups, the GDNF-treated group exhibited the highest expression of miR-21 (*P < 0.05). Caspase-3 expression was lower in the epididymosome-treated group than in the other groups (***P < 0.001). Compared to the 3D and negative control groups, the 3D + epididymosomes and 3D + GDNF groups showed an increase in spermatogenic cells. Immunohistochemical results confirmed the growth and differentiation of spermatogonial cells into spermatids in the treatment groups. CONCLUSION The DTM hydrogel containing 20 µg/mL epididymosomes or 10 ng/mL GDNF is a novel and safe culture system that can support SSC proliferation in vitro to obtain adequate SSCs for transplantation success. It could be a novel therapeutic agent that could recover deregulated SSCs in azoospermic patients.
Collapse
Affiliation(s)
- Maryam Rahbar
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Reza Asadpour
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Zohreh Mazaheri
- Basic Medical Science Research Center, Histogenotech Company, Tehran, Iran
| |
Collapse
|
4
|
Battistone MA, Elizagaray ML, Barrachina F, Ottino K, Mendelsohn AC, Breton S. Immunoregulatory mechanisms between epithelial clear cells and mononuclear phagocytes in the epididymis. Andrology 2024; 12:949-963. [PMID: 37572347 PMCID: PMC10859549 DOI: 10.1111/andr.13509] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/14/2023]
Abstract
INTRODUCTION One of the most intriguing aspects of male reproductive physiology is the ability of the epididymis to prevent the mounting of immune responses against the onslaught of foreign antigens carried by spermatozoa while initiating very efficient immune responses versus stressors. Epithelial clear cells are strategically positioned to work in a concerted manner with region-specific heterogeneous subsets of mononuclear phagocytes to survey the epididymal barrier and regulate the balance between inflammation and immune tolerance in the post-testicular environment. OBJECTIVE This review aims to describe how clear cells communicate with mononuclear phagocytes to contribute to the unique immune environment in which sperm mature and are stored in the epididymis. MATERIALS/METHODS A comprehensive systematic review was performed. PubMed was searched for articles specific to clear cells, mononuclear phagocytes, and epididymis. Articles that did not specifically address the target material were excluded. RESULTS In this review, we discuss the unexpected roles of clear cells, including the transfer of new proteins to spermatozoa via extracellular vesicles and nanotubes as they transit along the epididymal tubule; and we summarize the immune phenotype, morphology, and antigen capturing, processing, and presenting abilities of mononuclear phagocytes. Moreover, we present the current knowledge of immunoregulatory mechanisms by which clear cells and mononuclear phagocytes may contribute to the immune-privileged environment optimal for sperm maturation and storage. DISCUSSION AND CONCLUSION Notably, we provide an in-depth characterization of clear cell-mononuclear phagocyte communication networks in the steady-state epididymis and in the presence of injury. This review highlights crucial concepts of mucosal immunology and cellcell interactions, all of which are critical but understudied facets of human male reproductive health.
Collapse
Affiliation(s)
- MA Battistone
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - ML Elizagaray
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - F Barrachina
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - K Ottino
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - AC Mendelsohn
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - S Breton
- Centre Hospitalier Universitaire de Québec-Research Center, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Québec (Québec), Canada
| |
Collapse
|
5
|
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
|
6
|
Relovska S, Wang H, Zhang X, Fernández-Tussy P, Jeong KJ, Choi J, Suárez Y, McDonald JG, Fernández-Hernando C, Chung JJ. DHCR24-mediated sterol homeostasis during spermatogenesis is required for sperm mitochondrial sheath formation and impacts male fertility over time. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.21.572851. [PMID: 38187697 PMCID: PMC10769317 DOI: 10.1101/2023.12.21.572851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Desmosterol and cholesterol are essential lipid components of the sperm plasma membrane. Cholesterol efflux is required for capacitation, a process through which sperm acquire fertilizing ability. In this study, using a transgenic mouse model overexpressing 24-dehydrocholesterol reductase (DHCR24), an enzyme in the sterol biosynthesis pathway responsible for the conversion of desmosterol to cholesterol, we show that disruption of sterol homeostasis during spermatogenesis led to defective sperm morphology characterized by incomplete mitochondrial packing in the midpiece, reduced sperm count and motility, and a decline in male fertility with increasing paternal age, without changes in body fat composition. Sperm depleted of desmosterol exhibit inefficiency in the acrosome reaction, metabolic dysfunction, and an inability to fertilize the egg. These findings provide molecular insights into sterol homeostasis for sperm capacitation and its impact on male fertility.
Collapse
Affiliation(s)
- Sona Relovska
- 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
| | - Xinbo Zhang
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Pablo Fernández-Tussy
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Kyung Jo Jeong
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Yajaira Suárez
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Jeffrey G. McDonald
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Carlos Fernández-Hernando
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Pathology, Yale University 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 Gynecology and Obstetrics, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
7
|
Chen C, Zhang Z, Gu X, Sheng X, Xiao L, Wang X. Exosomes: New regulators of reproductive development. Mater Today Bio 2023; 19:100608. [PMID: 36969697 PMCID: PMC10034510 DOI: 10.1016/j.mtbio.2023.100608] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/12/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023] Open
Abstract
Exosomes are a subtype of extracellular vesicles (EVs) with a size range between 30 and 150 nm, which can be released by the majority of cell types and circulate in body fluid. They function as a long-distance cell-to-cell communication mechanism that modulates the gene expression profile and fate of target cells. Increasing evidence has indicated exosomes' central role in regulating various complex reproductive processes. However, to our knowledge, a review that focally and vividly describes the role of exosomes in reproductive development is still lacking. This review highlights our knowledge about the contribution of exosomes to early mammalian reproduction, such as gametogenesis, fertilization, early embryonic development, implantation, placentation and pregnancy. The discussion is primarily drawn from literature pertaining to the mammalian lineage with emphasis on the roles of exosomes in human reproduction and laboratory and livestock models.
Collapse
|
8
|
Belleannée C, Viana AGDA, Lavoie-Ouellet C. Intra and intercellular signals governing sperm maturation. Reprod Fertil Dev 2022; 35:27-38. [PMID: 36592975 DOI: 10.1071/rd22226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
After their production in the testis, spermatozoa do not have the capacity to move progressively and are unable to fertilise an oocyte. They sequentially acquire these abilities following their maturation in the epididymis and their capacitation/hyperactivation in the female reproductive system. As gene transcription is silenced in spermatozoa, extracellular factors released from the epididymal epithelium and from secretory glands allow spermatozoa to acquire bioactive molecules and to undergo intrinsic modifications. These modifications include epigenetic changes and post-translational modifications of endogenous proteins, which are important processes in sperm maturation. This article emphasises the roles played by extracellular factors secreted by the epididymis and accessory glands in the control of sperm intercellular signallings and fertilising abilities.
Collapse
Affiliation(s)
- Clémence Belleannée
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, Université Laval, Center for Research in Reproduction, Development and Intergenerational Health (CRDSI), CHU de Québec Research Center (CHUL), Quebec City, QC, Canada
| | | | - Camille Lavoie-Ouellet
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, Université Laval, Center for Research in Reproduction, Development and Intergenerational Health (CRDSI), CHU de Québec Research Center (CHUL), Quebec City, QC, Canada
| |
Collapse
|
9
|
Skerrett-Byrne DA, Anderson AL, Bromfield EG, Bernstein IR, Mulhall JE, Schjenken JE, Dun MD, Humphrey SJ, Nixon B. Global profiling of the proteomic changes associated with the post-testicular maturation of mouse spermatozoa. Cell Rep 2022; 41:111655. [DOI: 10.1016/j.celrep.2022.111655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 06/15/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022] Open
|
10
|
The Mechanism and Experimental Validation of Forsythoside A in the Treatment of Male Infertility Were Analyzed Based on Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7723358. [PMID: 36248414 PMCID: PMC9560825 DOI: 10.1155/2022/7723358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/15/2022]
Abstract
Chinese medicine extracts are currently the hotspot of new drug research and development. Herein, we report the mechanism of action of the traditional Chinese medicine extract Forsythiaside A in the treatment of male infertility and experimental verification. We first obtained 95 intersection genes between the target protein of Forsythiaside A and the target genes of male infertility and screened 13 key genes. In molecular docking, Forsythiaside A can each have a higher total docking score with 12 key genes and have a better combination. These 95 intersection genes are mainly related to biological processes such as response to peptide hormone, response to oxidative stress, and participation in the oxidative stress of the forkhead box O (FoxO) signaling pathway. Therefore, we use ornidazole to induce an experimental model of oligoasthenospermia in rats and use different concentrations of Forsythiaside A to intervene. We proved that the semen quality and superoxide dismutase (SOD) activities of model group rats were significantly lower than those of the blank group, and semen quality and SOD activities of the low-dose group and high-dose group were significantly higher than those of the model group. The malondialdehyde (MDA) level of model group rats was significantly higher than that of blank group, while the MDA levels of the low-dose group and high-dose group were significantly lower than that of the model group. Forsythoside A is a potential drug substance for male infertility and improves the semen quality, MDA levels, and SOD activities of rats with oligoasthenospermia.
Collapse
|
11
|
Cavariani MM, de Mello Santos T, Chuffa LGDA, Pinheiro PFF, Scarano WR, Domeniconi RF. Maternal Protein Restriction Alters the Expression of Proteins Related to the Structure and Functioning of the Rat Offspring Epididymis in an Age-Dependent Manner. Front Cell Dev Biol 2022; 10:816637. [PMID: 35517501 PMCID: PMC9061959 DOI: 10.3389/fcell.2022.816637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/05/2022] [Indexed: 11/16/2022] Open
Abstract
Nutrition is an environmental factor able to activate physiological interactions between fetus and mother. Maternal protein restriction is able to alter sperm parameters associated with epididymal functions. Since correct development and functioning of the epididymides are fundamental for mammalian reproductive success, this study investigated the effects of maternal protein restriction on epididymal morphology and morphometry in rat offspring as well as on the expression of Src, Cldn-1, AR, ER, aromatase p450, and 5α-reductase in different stages of postnatal epididymal development. For this purpose, pregnant females were allocated to normal-protein (NP—17% protein) and low-protein (LP—6% protein) groups that received specific diets during gestation and lactation. After weaning, male offspring was provided only normal-protein diet until the ages of 21, 44, and 120 days, when they were euthanized and their epididymides collected. Maternal protein restriction decreased genital organs weight as well as crown-rump length and anogenital distance at all ages. Although the low-protein diet did not change the integrity of the epididymal epithelium, we observed decreases in tubular diameter, epithelial height and luminal diameter of the epididymal duct in 21-day-old LP animals. The maternal low-protein diet changed AR, ERα, ERβ, Src 416, and Src 527 expression in offspring epididymides in an age-dependent manner. Finally, maternal protein restriction increased Cldn-1 expression throughout the epididymides at all analyzed ages. Although some of these changes did not remain until adulthood, the insufficient supply of proteins in early life altered the structure and functioning of the epididymis in important periods of postnatal development.
Collapse
|
12
|
Barrachina F, Battistone MA, Castillo J, Mallofré C, Jodar M, Breton S, Oliva R. Sperm acquire epididymis-derived proteins through epididymosomes. Hum Reprod 2022; 37:651-668. [PMID: 35137089 PMCID: PMC8971652 DOI: 10.1093/humrep/deac015] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 12/30/2021] [Indexed: 02/07/2023] Open
Abstract
STUDY QUESTION Are epididymosomes implicated in protein transfer from the epididymis to spermatozoa? SUMMARY ANSWER We characterized the contribution of epididymal secretions to the sperm proteome and demonstrated that sperm acquire epididymal proteins through epididymosomes. WHAT IS KNOWN ALREADY Testicular sperm are immature cells unable to fertilize an oocyte. After leaving the testis, sperm transit along the epididymis to acquire motility and fertilizing abilities. It is well known that marked changes in the sperm proteome profile occur during epididymal maturation. Since the sperm is a transcriptional and translational inert cell, previous studies have shown that sperm incorporate proteins, RNA and lipids from extracellular vesicles (EVs), released by epithelial cells lining the male reproductive tract. STUDY DESIGN, SIZE, DURATION We examined the contribution of the epididymis to the post-testicular maturation of spermatozoa, via the production of EVs named epididymosomes, released by epididymal epithelial cells. An integrative analysis using both human and mouse data was performed to identify sperm proteins with a potential epididymis-derived origin. Testes and epididymides from adult humans (n = 9) and adult mice (n = 3) were used to experimentally validate the tissue localization of four selected proteins using high-resolution confocal microscopy. Mouse epididymal sperm were co-incubated with carboxyfluorescein succinimidyl ester (CFSE)-labeled epididymosomes (n = 4 mice), and visualized using high-resolution confocal microscopy. PARTICIPANTS/MATERIALS, SETTING, METHODS Adult (12-week-old) C57BL/CBAF1 wild-type male mice and adult humans were used for validation purposes. Testes and epididymides from both mice and humans were obtained and processed for immunofluorescence. Mouse epididymal sperm and mouse epididymosomes were obtained from the epididymal cauda segment. Fluorescent epididymosomes were obtained after labeling the epididymal vesicles with CFSE dye followed by epididymosome isolation using a density cushion. Immunofluorescence was performed following co-incubation of sperm with epididymosomes in vitro. High-resolution confocal microscopy and 3D image reconstruction were used to visualize protein localization and sperm-epididymosomes interactions. MAIN RESULTS AND THE ROLE OF CHANCE Through in silico analysis, we first identified 25 sperm proteins with a putative epididymal origin that were conserved in both human and mouse spermatozoa. From those, the epididymal origin of four sperm proteins (SLC27A2, EDDM3B, KRT19 and WFDC8) was validated by high-resolution confocal microscopy. SLC27A2, EDDM3B, KRT19 and WFDC8 were all detected in epithelial cells lining the human and mouse epididymis, and absent from human and mouse seminiferous tubules. We found region-specific expression patterns of these proteins throughout the mouse epididymides. In addition, while EDDM3B, KRT19 and WFDC8 were detected in both epididymal principal and clear cells (CCs), SLC27A2 was exclusively expressed in CCs. Finally, we showed that CFSE-fluorescently labeled epididymosomes interact with sperm in vitro and about 12-36% of the epididymosomes contain the targeted sperm proteins with an epididymal origin. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The human and mouse sample size was limited and our results were descriptive. The analyses of epididymal sperm and epididymosomes were solely performed in the mouse model due to the difficulties in obtaining epididymal luminal fluid human samples. Alternatively, human ejaculated sperm and seminal EVs could not be used because ejaculated sperm have already contacted with the fluids secreted by the male accessory sex glands, and seminal EVs contain other EVs in addition to epididymosomes, such as the abundant prostate-derived EVs. WIDER IMPLICATIONS OF THE FINDINGS Our findings indicate that epididymosomes are capable of providing spermatozoa with a new set of epididymis-derived proteins that could modulate the sperm proteome and, subsequently, participate in the post-testicular maturation of sperm cells. Additionally, our data provide further evidence of the novel role of epididymal CCs in epididymosome production. Identifying mechanisms by which sperm mature to acquire their fertilization potential would, ultimately, lead to a better understanding of male reproductive health and may help to identify potential therapeutic strategies to improve male infertility. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the Spanish Ministry of Economy and Competitiveness (Ministerio de Economía y Competividad; fondos FEDER 'una manera de hacer Europa' PI13/00699 and PI16/00346 to R.O.; and Sara Borrell Postdoctoral Fellowship, Acción Estratégica en Salud, CD17/00109 to J.C.), by National Institutes of Health (grants HD040793 and HD069623 to S.B., grant HD104672-01 to M.A.B.), by the Spanish Ministry of Education, Culture and Sports (Ministerio de Educación, Cultura y Deporte para la Formación de Profesorado Universitario, FPU15/02306 to F.B.), by a Lalor Foundation Fellowship (to F.B. and M.A.B.), by the Government of Catalonia (Generalitat de Catalunya, pla estratègic de recerca i innovació en salut, PERIS 2016-2020, SLT002/16/00337 to M.J.), by Fundació Universitària Agustí Pedro i Pons (to F.B.), and by the American Society for Biochemistry and Molecular Biology (PROLAB Award from ASBMB/IUBMB/PABMB to F.B.). Confocal microscopy and transmission electron microscopy was performed in the Microscopy Core facility of the Massachusetts General Hospital (MGH) Center for Systems Biology/Program in Membrane Biology which receives support from Boston Area Diabetes and Endocrinology Research Center (BADERC) award DK57521 and Center for the Study of Inflammatory Bowel Disease grant DK43351. The Zeiss LSM800 microscope was acquired using an NIH Shared Instrumentation Grant S10-OD-021577-01. The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- F Barrachina
- Molecular Biology of Reproduction and Development Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - M A Battistone
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - J Castillo
- Molecular Biology of Reproduction and Development Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - C Mallofré
- Department of Pathology, Universitat de Barcelona, Hospital Clínic, Barcelona, Spain
| | - M Jodar
- Molecular Biology of Reproduction and Development Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic, Barcelona, Spain
| | - S Breton
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - R Oliva
- Molecular Biology of Reproduction and Development Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Fundació Clínic per a la Recerca Biomèdica, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic, Barcelona, Spain
| |
Collapse
|
13
|
Chen H, Scott-Boyer MP, Droit A, Robert C, Belleannée C. Sperm Heterogeneity Accounts for Sperm DNA Methylation Variations Observed in the Caput Epididymis, Independently From DNMT/TET Activities. Front Cell Dev Biol 2022; 10:834519. [PMID: 35392175 PMCID: PMC8981467 DOI: 10.3389/fcell.2022.834519] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/04/2022] [Indexed: 11/15/2022] Open
Abstract
Following their production in the testis, spermatozoa enter the epididymis where they gain their motility and fertilizing abilities. This post-testicular maturation coincides with sperm epigenetic profile changes that influence progeny outcome. While recent studies highlighted the dynamics of small non-coding RNAs in maturing spermatozoa, little is known regarding sperm methylation changes and their impact at the post-fertilization level. Fluorescence-activated cell sorting (FACS) was used to purify spermatozoa from the testis and different epididymal segments (i.e., caput, corpus and cauda) of CAG/su9-DsRed2; Acr3-EGFP transgenic mice in order to map out sperm methylome dynamics. Reduced representation bisulfite sequencing (RRBS-Seq) performed on DNA from these respective sperm populations indicated that high methylation changes were observed between spermatozoa from the caput vs. testis with 5,546 entries meeting our threshold values (q value <0.01, methylation difference above 25%). Most of these changes were transitory during epididymal sperm maturation according to the low number of entries identified between spermatozoa from cauda vs. testis. According to enzymatic and sperm/epididymal fluid co-incubation assays, (de)methylases were not found responsible for these sperm methylation changes. Instead, we identified that a subpopulation of caput spermatozoa displayed distinct methylation marks that were susceptible to sperm DNAse treatment and accounted for the DNA methylation profile changes observed in the proximal epididymis. Our results support the paradigm that a fraction of caput spermatozoa has a higher propensity to bind extracellular DNA, a phenomenon responsible for the sperm methylome variations observed at the post-testicular level. Further investigating the degree of conservation of this sperm heterogeneity in human will eventually provide new considerations regarding sperm selection procedures used in fertility clinics.
Collapse
Affiliation(s)
- Hong Chen
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
- Center for Research in Reproduction, Development and Intergenerational Health, Quebec, QC, Canada
| | | | - Arnaud Droit
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - Claude Robert
- Center for Research in Reproduction, Development and Intergenerational Health, Quebec, QC, Canada
- Faculty of Animal Sciences, Université Laval, Quebec, QC, Canada
| | - Clémence Belleannée
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
- Center for Research in Reproduction, Development and Intergenerational Health, Quebec, QC, Canada
- *Correspondence: Clémence Belleannée,
| |
Collapse
|
14
|
An Update on Semen Physiology, Technologies, and Selection Techniques for the Advancement of In Vitro Equine Embryo Production: Section I. Animals (Basel) 2021; 11:ani11113248. [PMID: 34827983 PMCID: PMC8614440 DOI: 10.3390/ani11113248] [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: 10/09/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/05/2022] Open
Abstract
Simple Summary Male fertility is often estimated by simple sperm assessment, and therefore, it is crucial to establish species-specific baselines for normal sperm parameters. In this paper, sperm physiology, function, and common abnormalities in stallions will be reviewed. Abstract As the use of assisted reproductive technologies (ART) and in vitro embryo production (IVP) expand in the equine industry, it has become necessary to further our understanding of semen physiology as it applies to overall fertility. This segment of our two-section review will focus on normal sperm parameters, beginning with development and extending through the basic morphology of mature spermatozoa, as well as common issues with male factor infertility in IVP. Ultimately, the relevance of sperm parameters to overall male factor fertility in equine IVP will be assessed.
Collapse
|
15
|
Chen H, Alves MBR, Belleannée C. Contribution of epididymal epithelial cell functions to sperm epigenetic changes and the health of progeny. Hum Reprod Update 2021; 28:51-66. [PMID: 34618012 DOI: 10.1093/humupd/dmab029] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/19/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Spermatozoa acquire their motility and fertilizing abilities during their maturation through the epididymis. This process is controlled by epididymal epithelial cells that possess features adapted to sense and respond to their surrounding environment and to communicate with spermatozoa. During the past decade, new intercellular communication processes have been discovered, including the secretion and transport of molecules from the epithelium to spermatozoa via extracellular vesicles (EVs), as well as sensing of the intraluminal milieu by cellular extensions. OBJECTIVE AND RATIONALE This review addresses recent findings regarding epididymal epithelial cell features and interactions between spermatozoa and the epididymal epithelium as well as epigenetic modifications undergone by spermatozoa during transit through the epididymal microenvironment. SEARCH METHODS A systematic search was conducted in Pubmed with the keyword 'epididymis'. Results were filtered on original research articles published from 2009 to 2021 and written in the English language. One hundred fifteen original articles presenting recent advancements on the epididymis contribution to sperm maturation were selected. Some additional papers cited in the primary reference were also included. A special focus was given to higher mammalian species, particularly rodents, bovines and humans, that are the most studied in this field. OUTCOMES This review provides novel insights into the contribution of epididymal epithelium and EVs to post-testicular sperm maturation. First, new immune cell populations have been described in the epididymis, where they are proposed to play a role in protecting the environment surrounding sperm against infections or autoimmune responses. Second, novel epididymal cell extensions, including dendrites, axopodia and primary cilia, have been identified as sensors of the environment surrounding sperm. Third, new functions have been outlined for epididymal EVs, which modify the sperm epigenetic profile and participate in transgenerational epigenetic inheritance of paternal traits. WIDER IMPLICATIONS Although the majority of these findings result from studies in rodents, this fundamental research will ultimately improve our knowledge of human reproductive physiopathologies. Recent discoveries linking sperm epigenetic modifications with paternal environmental exposure and progeny outcome further stress the importance of advancing fundamental research on the epididymis. From this, new therapeutic options for infertile couples and better counseling strategies may arise to increase positive health outcomes in children conceived either naturally or with ART.
Collapse
Affiliation(s)
- Hong Chen
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, Quebec, Canada
| | | | - Clémence Belleannée
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, Quebec, Canada
| |
Collapse
|
16
|
Tourzani DA, Battistone MA, Salicioni AM, Breton S, Visconti PE, Gervasi MG. Caput Ligation Renders Immature Mouse Sperm Motile and Capable to Undergo cAMP-Dependent Phosphorylation. Int J Mol Sci 2021; 22:ijms221910241. [PMID: 34638585 PMCID: PMC8549708 DOI: 10.3390/ijms221910241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/01/2022] Open
Abstract
Mammalian sperm must undergo two post-testicular processes to become fertilization-competent: maturation in the male epididymis and capacitation in the female reproductive tract. While caput epididymal sperm are unable to move and have not yet acquired fertilization potential, sperm in the cauda epididymis have completed their maturation, can move actively, and have gained the ability to undergo capacitation in the female tract or in vitro. Due to the impossibility of mimicking sperm maturation in vitro, the molecular pathways underlying this process remain largely unknown. We aimed to investigate the use of caput epididymal ligation as a tool for the study of sperm maturation in mice. Our results indicate that after seven days of ligation, caput sperm gained motility and underwent molecular changes comparable with those observed for cauda mature sperm. Moreover, ligated caput sperm were able to activate pathways related to sperm capacitation. Despite these changes, ligated caput sperm were unable to fertilize in vitro. Our results suggest that transit through the epididymis is not required for the acquisition of motility and some capacitation-associated signaling but is essential for full epididymal maturation. Caput epididymal ligation is a useful tool for the study of the molecular pathways involved in the acquisition of sperm motility during maturation.
Collapse
Affiliation(s)
- Darya A. Tourzani
- Department of Veterinary and Animal Sciences, Integrated Science Building, University of Massachusetts, Amherst, MA 01003, USA; (D.A.T.); (A.M.S.)
| | - Maria A. Battistone
- Program in Membrane Biology, Nephrology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA; (M.A.B.); (S.B.)
| | - Ana M. Salicioni
- Department of Veterinary and Animal Sciences, Integrated Science Building, University of Massachusetts, Amherst, MA 01003, USA; (D.A.T.); (A.M.S.)
| | - Sylvie Breton
- Program in Membrane Biology, Nephrology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA; (M.A.B.); (S.B.)
| | - Pablo E. Visconti
- Department of Veterinary and Animal Sciences, Integrated Science Building, University of Massachusetts, Amherst, MA 01003, USA; (D.A.T.); (A.M.S.)
- Correspondence: (P.E.V.); (M.G.G.); Tel.: +1-413-545-5565 (P.E.V.)
| | - Maria G. Gervasi
- Department of Veterinary and Animal Sciences, Integrated Science Building, University of Massachusetts, Amherst, MA 01003, USA; (D.A.T.); (A.M.S.)
- Correspondence: (P.E.V.); (M.G.G.); Tel.: +1-413-545-5565 (P.E.V.)
| |
Collapse
|
17
|
Abstract
More than a century ago, August Weissman defined a distinction between the germline (responsible for propagating heritable information from generation to generation) and the perishable soma. A central motivation for this distinction was to argue against the inheritance of acquired characters, as the germline was partly defined by its protection from external conditions. However, recent decades have seen an explosion of studies documenting the intergenerational and transgenerational effects of environmental conditions, forcing a re-evaluation of how external signals are sensed by, or communicated to, the germline epigenome. Here, motivated by the centrality of small RNAs in paradigms of epigenetic inheritance, we review across species the myriad examples of intercellular RNA trafficking from nurse cells or somatic tissues to developing gametes.
Collapse
|
18
|
Kim SW, Kim B. Vacuolar H(+)-ATPase is not restricted to clear cells of the epididymal epithelium in cattle. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2021; 63:262-271. [PMID: 33987602 PMCID: PMC8071741 DOI: 10.5187/jast.2021.e32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 11/28/2022]
Abstract
Communication among epididymal epithelial cells creates the best luminal
condition where spermatozoa mature, transport and are stored. Vacuolar ATPase
(V-ATPase) and cytokeratin 5 (KRT5) have been used as signal indicators for
clear and basal cells of the epididymal epithelium, respectively, in mice, rats,
bats, and pigs; however, these two markers have not yet been described in the
epididymis of bulls. Here, we examined the presence and distribution of the B1
subunit of V-ATPase (B1-VATPase) and KRT5 in the distinct regions of adult
bovine epididymides, specifically, the caput, corpus, and cauda.
Immunofluorescence staining and confocal microscopy showed that narrow
shaped-clear cells were placed in the caput and corpus regions of the bovine
epididymis; however, they were absent in the cauda epididymis. In addition,
B1-VATPase was highly expressed in the cauda spermatozoa; however, it was rarely
detected in the caput spermatozoa. On the other hand, KRT5-positive cells, basal
cells, were maintained beneath the basal lamina and they had the traditional
form with a dome-shaped morphology from the caput to cauda region of the bovine
epididymis. The co-expression of B1-VATPase and KRT5 was confined to basal cells
placed in the basal region of the epithelium. In summary, 1) clear cells were
present with region-specific localization, 2) B1-VATPase was present in the
corpus and cauda spermatozoa but absent in the caput, 3) co-expressed cells with
B1-VATPase and KRT5 were present in the adult bovine epididymis, and 4)
B1-VATPase was not a specific marker for clear cells in the bovine epididymis.
Therefore, the perfect epididymal luminal condition created by the specific
expression and localization patterns of B1-VATPase might be necessary to obtain
fertilizing capacity of spermatozoa in the bovine epididymis.
Collapse
Affiliation(s)
- Sung Woo Kim
- Animal Genetic Resource Research Center, National Institute of Animal Science, Hamyang 50000, Korea
| | - Bongki Kim
- Department of Animal Resources Science, Kongju National University, Yesan 32439, Korea
| |
Collapse
|
19
|
Galan C, Serra RW, Sun F, Rinaldi VD, Conine CC, Rando OJ. Stability of the cytosine methylome during post-testicular sperm maturation in mouse. PLoS Genet 2021; 17:e1009416. [PMID: 33661909 PMCID: PMC7963034 DOI: 10.1371/journal.pgen.1009416] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/16/2021] [Accepted: 02/14/2021] [Indexed: 12/28/2022] Open
Abstract
Beyond the haploid genome, mammalian sperm carry a payload of epigenetic information with the potential to modulate offspring phenotypes. Recent studies show that the small RNA repertoire of sperm is remodeled during post-testicular maturation in the epididymis. Epididymal maturation has also been linked to changes in the sperm methylome, suggesting that the epididymis might play a broader role in shaping the sperm epigenome. Here, we characterize the genome-wide methylation landscape in seven germ cell populations from throughout the male reproductive tract. We find very few changes in the cytosine methylation landscape between testicular germ cell populations and cauda epididymal sperm, demonstrating that the sperm methylome is stable throughout post-testicular maturation. Although our sequencing data suggested that caput epididymal sperm exhibit a highly unusual methylome, follow-up studies revealed that this resulted from contamination of caput sperm by extracellular DNA. Extracellular DNA formed web-like structures that ensnared sperm, and was present only in sperm samples obtained from the caput epididymis and vas deferens of virgin males. Curiously, contaminating extracellular DNA was associated with citrullinated histone H3, potentially resulting from a PAD-driven genome decondensation process. Taken together, our data emphasize the stability of cytosine methylation in mammalian sperm, and identify a surprising, albeit transient, period during which sperm are associated with extracellular DNA.
Collapse
Affiliation(s)
- Carolina Galan
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Ryan W. Serra
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Fengyun Sun
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Vera D. Rinaldi
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Colin C. Conine
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Oliver J. Rando
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
20
|
Zhang P, Huang Y, Fu Q, He W, Xiao K, Zhang M. Integrated analysis of phosphoproteome and ubiquitylome in epididymal sperm of buffalo (Bubalus bubalis). Mol Reprod Dev 2021; 88:15-33. [PMID: 33140506 PMCID: PMC7894524 DOI: 10.1002/mrd.23432] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/14/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022]
Abstract
In mammals, sperm need to mature in the epididymis to gain fertilization competency. However, the molecular mechanism underlying buffalo sperm maturation remains elusive. Exploring sperm physiology at the posttranslational modification (PTM) level could help to develop our understanding of these mechanisms. Protein phosphorylation and ubiquitination are major PTMs in the regulation of many biological processes. In the present study, to our knowledge, we report the first phosphoproteome and ubiquitylome of sperm collected from the caput, corpus, and cauda segments of the epididymis using liquid chromatography-mass spectrometry combined with affinity purification. In total, 647 phosphorylation sites in 294 proteins and 1063 ubiquitination sites in 446 proteins were characterized. Some of these proteins were associated with cellular developmental processes and energy metabolic pathways. Interestingly, 84 proteins were both phosphorylated and ubiquitinated, simultaneously. Some of these proteins were involved in, for example, spermatogenesis, reproduction, and spermatid development. Taken together, these data provide a theoretical basis for further functional analysis of phosphorylation and ubiquitination in epididymal sperm of buffalo and other mammals, and serve as an important resource for exploring the physiological mechanism underlying sperm maturation.
Collapse
Affiliation(s)
- Peng‐fei Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources, Animal Reproduction InstituteGuangxi UniversityNanningGuangxiChina
| | - Yu‐lin Huang
- Department of Cell and Genetics, College of Basic MedicineGuangxi University of Chinese MedicineNanningGuangxiChina
| | - Qiang Fu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources, Animal Reproduction InstituteGuangxi UniversityNanningGuangxiChina
| | - Weng‐tan He
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources, Animal Reproduction InstituteGuangxi UniversityNanningGuangxiChina
| | - Kai Xiao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources, Animal Reproduction InstituteGuangxi UniversityNanningGuangxiChina
| | - Ming Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources, Animal Reproduction InstituteGuangxi UniversityNanningGuangxiChina
| |
Collapse
|
21
|
Abstract
Within the reproductive tract, distinct cell types must have precisely controlled communication for complex processes such as gamete production, fertilisation and implantation. Intercellular communication in many physiological processes involves extracellular vesicles (EVs). In reproductive systems, EVs have been implicated in many aspects, from gamete maturation to embryo development. Sperm develop within the testis and then exit into the epididymis in an immature form, lacking motility and fertilising capabilities. Due to their small size, compact nature of the nucleus and the lack of specific organelles, sperm are unable to perform de novo protein synthesis, and thus rely on extrinsic signals delivered from the external milieu to gain full function. Mounting evidence points to EVs as being a major provider of these signals, not just within the male reproductive tract but also within the female as the sperm make their way through a seemingly hostile environment to the oocyte. In this chapter, we review the current knowledge on EVs as mediators of sperm maturation and function and highlight their potential roles in male fertility.
Collapse
Affiliation(s)
- Natalie J Foot
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| |
Collapse
|
22
|
Pujianto D, Permatasari S. Mouse CD52 is predominantly expressed in the cauda epididymis, regulated by androgen and lumicrine factors. J Hum Reprod Sci 2021; 14:350-355. [PMID: 35197679 PMCID: PMC8812399 DOI: 10.4103/jhrs.jhrs_29_21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 08/10/2021] [Accepted: 12/02/2021] [Indexed: 11/12/2022] Open
Abstract
Background: Sperm maturation takes place through contact between sperm and proteins produced in the epididymal lumen. CD52 had been characterised in the sperm; however, the expression and its regulation in the epididymis are mostly unknown. Aim: This study aimed to analyse the expression and regulation of CD52 in the mouse epididymis. Setting and Design: Experimental design was used in this study. Materials and Methods: Epididymis tissues from mice strain Deutch Democratic Yokohama were used as sources of total RNA. Bioinformatic tool was used to predict signal peptides. Quantitative real-time reverse transcription–polymerase chain reaction was used to analyse tissue distribution, androgen, testicular factors dependency and postnatal development. Statistical Analysis: One-way analysis of variance was used to analyse differences between treatment and control untreated group. P < 0.05 was determined as a significant difference. Results: CD52 amino acid sequence contains a signal peptide, indicating it is a secretory protein. CD52 exhibited region-specific expression in the epididymis, with the highest level being in the cauda. CD52 expression was regulated by androgen indicated by a significant downregulation at day 1 and day 3 following a castration (P < 0.05). Dependency on androgen was confirmed by injection of exogenous testosterone which prevented downregulation by 50%. Moreover, lumicrine factors also influenced CD52 expression indicated by ligation of efferent duct which also reduced expression at day 1 to day 5 following the ligation (P < 0.05). CD52 expression was developmentally regulated. This was shown by increase in the level of expression starting at day 15 postnatally. Conclusion: CD52 shows characteristics of genes involved in sperm maturation in the epididymis.
Collapse
|
23
|
Rinaldi VD, Donnard E, Gellatly K, Rasmussen M, Kucukural A, Yukselen O, Garber M, Sharma U, Rando OJ. An atlas of cell types in the mouse epididymis and vas deferens. eLife 2020; 9:e55474. [PMID: 32729827 PMCID: PMC7426093 DOI: 10.7554/elife.55474] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Following testicular spermatogenesis, mammalian sperm continue to mature in a long epithelial tube known as the epididymis, which plays key roles in remodeling sperm protein, lipid, and RNA composition. To understand the roles for the epididymis in reproductive biology, we generated a single-cell atlas of the murine epididymis and vas deferens. We recovered key epithelial cell types including principal cells, clear cells, and basal cells, along with associated support cells that include fibroblasts, smooth muscle, macrophages and other immune cells. Moreover, our data illuminate extensive regional specialization of principal cell populations across the length of the epididymis. In addition to region-specific specialization of principal cells, we find evidence for functionally specialized subpopulations of stromal cells, and, most notably, two distinct populations of clear cells. Our dataset extends on existing knowledge of epididymal biology, and provides a wealth of information on potential regulatory and signaling factors that bear future investigation.
Collapse
Affiliation(s)
- Vera D Rinaldi
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Elisa Donnard
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Kyle Gellatly
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Morten Rasmussen
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Alper Kucukural
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Onur Yukselen
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Manuel Garber
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Medical SchoolWorcesterUnited States
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Upasna Sharma
- Department of Molecular, Cell and Developmental Biology, University of California Santa CruzSanta CruzUnited States
| | - Oliver J Rando
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical SchoolWorcesterUnited States
| |
Collapse
|
24
|
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
|
25
|
Hippo kinases MST1 and MST2 control the differentiation of the epididymal initial segment via the MEK-ERK pathway. Cell Death Differ 2020; 27:2797-2809. [PMID: 32332916 DOI: 10.1038/s41418-020-0544-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 11/08/2022] Open
Abstract
Although the roles of the Hippo pathway in organogenesis and tumorigenesis have been well studied in multiple organs, its role in sperm maturation and male fertility has not been investigated. The initial segment (IS) of the epididymis plays a critical role in sperm maturation. IS differentiation is governed by ERK1/2, but the mechanisms of ERK1/2 activation in IS are not fully understood. Here we show that double knockout (dKO) of mammalian sterile 20-like kinases 1 and 2 (Mst1 and Mst2), homologs of Hippo in Drosophila, in the epididymal epithelium led to male infertility in mice. Sperm in the cauda epididymides of mutant mice were immotile with flagellar angulation and severely disorganized structures. Loss of Mst1/2 activated YAP and increased proliferation and cell death in all the segments of epididymis. The mutant mice showed substantially suppressed MEK/ERK signaling in the IS and failed IS differentiation. Deletion of Yap restored the reduced MEK/ERK signaling, and partially rescued the defective IS differentiation and fertility in Mst1/2 dKO mice. Our results demonstrate that YAP inhibits the MEK/ERK pathway in IS epithelial cells, and MST1/2 control IS differentiation and fertility at least partially by repressing YAP. Taken together, the Hippo pathway is essential for sperm maturation and male fertility.
Collapse
|
26
|
Battistone MA, Spallanzani RG, Mendelsohn AC, Capen D, Nair AV, Brown D, Breton S. Novel role of proton-secreting epithelial cells in sperm maturation and mucosal immunity. J Cell Sci 2019; 133:jcs.233239. [PMID: 31636115 DOI: 10.1242/jcs.233239] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022] Open
Abstract
Epithelial cells are immune sensors and mediators that constitute the first line of defense against infections. Using the epididymis, a model for studying tubular organs, we uncovered a novel and unexpected role for professional proton-secreting 'clear cells' in sperm maturation and immune defense. The epididymal epithelium participates in the maturation of spermatozoa via the establishment of an acidic milieu and transfer of proteins to sperm cells, a poorly characterized process. We show that proton-secreting clear cells express mRNA transcripts and proteins that are acquired by maturing sperm, and that they establish close interactions with luminal spermatozoa via newly described 'nanotubes'. Mechanistic studies show that injection of bacterial antigens in vivo induces chemokine expression in clear cells, followed by macrophage recruitment into the organ. Injection of an inflammatory intermediate mediator (IFN-γ) increased Cxcl10 expression in clear cells, revealing their participation as sensors and mediators of inflammation. The functional diversity adopted by clear cells might represent a generalized phenomenon by which similar epithelial cells decode signals, communicate with neighbors and mediate mucosal immunity, depending on their precise location within an organ.
Collapse
Affiliation(s)
- Maria A Battistone
- Program in Membrane Biology, Center for Systems Biology and Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Raul German Spallanzani
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alexandra C Mendelsohn
- Program in Membrane Biology, Center for Systems Biology and Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Diane Capen
- Program in Membrane Biology, Center for Systems Biology and Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Anil V Nair
- Program in Membrane Biology, Center for Systems Biology and Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Dennis Brown
- Program in Membrane Biology, Center for Systems Biology and Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Sylvie Breton
- Program in Membrane Biology, Center for Systems Biology and Nephrology Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| |
Collapse
|
27
|
Chu C, Zhang YL, Yu L, Sharma S, Fei ZL, Drevet JR. Epididymal small non-coding RNA studies: progress over the past decade. Andrology 2019; 7:681-689. [PMID: 31044548 DOI: 10.1111/andr.12639] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/01/2019] [Accepted: 03/30/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Small non-coding RNAs (sncRNAs) accomplish a huge variety of biological functions. Over the past decade, we have witnessed the substantial progress in the epididymal sncRNA studies. In the Epididymis 7, we had the true privilege of having a whole session to share our findings and exchange ideas on the epididymal sncRNA studies. OBJECTIVES This mini-review attempts to provide an overview of what is known about the sncRNAs in the mammalian epididymis and discuss the future directions in this field. METHODS We surveyed literature regarding the sncRNA studies in the mammalian epididymis, and integrated some of our unpublished findings as well. We focus on the progress in methodology and the advances in our understanding of the expression and functions of epididymal sncRNAs. RESULTS AND DISCUSSION The applications of high-throughput approaches have made great contributions in the discovery of new sncRNA species and profiling their dynamics in the epithelial cells, the passing spermatozoa, and the luminal environment. The diverse classes of epididymal sncRNAs exert important biological functions from the in situ regulation of epididymal gene expression to the epigenetic inheritance in the offspring. CONCLUSION Although still in its infancy, we believe that the research on epididymal sncRNAs will not only lead to a better understanding of their physiological and pathological functions, but also contribute to the whole landscape of the RNA field.
Collapse
Affiliation(s)
- C Chu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Y L Zhang
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - L Yu
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - S Sharma
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Z L Fei
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - J R Drevet
- Genetics Reproduction & Development Laboratory, CNRS UMR 6293 - INSERM U1103 - Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
28
|
Raspa M, Mahabir E, Paoletti R, Protti M, Mercolini L, Schiller P, Scavizzi F. Effects of oral d-aspartate on sperm quality in B6N mice. Theriogenology 2018; 121:53-61. [DOI: 10.1016/j.theriogenology.2018.07.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 07/30/2018] [Accepted: 07/30/2018] [Indexed: 11/16/2022]
|
29
|
T cell microvilli constitute immunological synaptosomes that carry messages to antigen-presenting cells. Nat Commun 2018; 9:3630. [PMID: 30194420 PMCID: PMC6128830 DOI: 10.1038/s41467-018-06090-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/17/2018] [Indexed: 12/22/2022] Open
Abstract
Microvilli on T cells have been proposed to survey surfaces of antigen-presenting cells (APC) or facilitate adhesion under flow; however, whether they serve essential functions during T cell activation remains unclear. Here we show that antigen-specific T cells deposit membrane particles derived from microvilli onto the surface of cognate antigen-bearing APCs. Microvilli carry T cell receptors (TCR) at all stages of T cell activation and are released as large TCR-enriched, T cell microvilli particles (TMP) in a process of trogocytosis. These microvilli exclusively contain protein arrestin-domain-containing protein 1, which is directly involved in membrane budding and, in combination with vacuolar protein-sorting-associated protein 4, transforms large TMPs into smaller, exosome-sized TMPs. Notably, TMPs from CD4+ T cells are enriched with LFA-2/CD2 and various cytokines involved in activating dendritic cells. Collectively, these results demonstrate that T cell microvilli constitute “immunological synaptosomes” that carry T cell messages to APCs. Microvilli can participate in adhesion or migration of T cells, but whether they are involved in function regulation is unclear. Here the authors show that T cell microvilli form budding vesicles containing T cell signalling components for deposition onto antigen presenting cells (APC) and modulation of APC functions.
Collapse
|
30
|
Sharma U, Sun F, Conine CC, Reichholf B, Kukreja S, Herzog VA, Ameres SL, Rando OJ. Small RNAs Are Trafficked from the Epididymis to Developing Mammalian Sperm. Dev Cell 2018; 46:481-494.e6. [PMID: 30057273 DOI: 10.1016/j.devcel.2018.06.023] [Citation(s) in RCA: 261] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 05/11/2018] [Accepted: 06/26/2018] [Indexed: 12/30/2022]
Abstract
The biogenesis of the RNA payload of mature sperm is of great interest, because RNAs delivered to the zygote at fertilization can affect early development. Here, we tested the hypothesis that small RNAs are trafficked to mammalian sperm during the process of post-testicular maturation in the epididymis. By characterizing small RNA dynamics during germ cell maturation in mice, we confirm and extend prior observations that sperm undergo a dramatic switch in the RNA payload from piRNAs to tRNA fragments (tRFs) upon exiting the testis and entering the epididymis. Small RNA delivery to sperm could be recapitulated in vitro by incubating testicular spermatozoa with caput epididymosomes. Finally, tissue-specific metabolic labeling of RNAs in intact mice definitively shows that mature sperm carry RNAs that were originally synthesized in the epididymal epithelium. These data demonstrate that soma-germline RNA transfer occurs in male mammals, most likely via vesicular transport from the epididymis to maturing sperm.
Collapse
Affiliation(s)
- Upasna Sharma
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Fengyun Sun
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Colin C Conine
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Brian Reichholf
- Institute of Molecular Biotechnology (IMBA), Vienna Biocenter Campus (VBC), 1030 Vienna, Austria
| | - Shweta Kukreja
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Veronika A Herzog
- Institute of Molecular Biotechnology (IMBA), Vienna Biocenter Campus (VBC), 1030 Vienna, Austria
| | - Stefan L Ameres
- Institute of Molecular Biotechnology (IMBA), Vienna Biocenter Campus (VBC), 1030 Vienna, Austria
| | - Oliver J Rando
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
31
|
Conine CC, Sun F, Song L, Rivera-Pérez JA, Rando OJ. Small RNAs Gained during Epididymal Transit of Sperm Are Essential for Embryonic Development in Mice. Dev Cell 2018; 46:470-480.e3. [PMID: 30057276 DOI: 10.1016/j.devcel.2018.06.024] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/03/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022]
Abstract
The small RNA payload of mammalian sperm undergoes dramatic remodeling during development, as several waves of microRNAs and tRNA fragments are shipped to sperm during post-testicular maturation in the epididymis. Here, we take advantage of this developmental process to probe the function of the sperm RNA payload in preimplantation development. We generated zygotes via intracytoplasmic sperm injection (ICSI) using sperm obtained from the proximal (caput) versus distal (cauda) epididymis and then characterized the development of the resulting embryos. Embryos generated using caput sperm significantly overexpress multiple regulatory factors throughout preimplantation development, subsequently implant inefficiently, and fail soon after implantation. Remarkably, microinjection of purified cauda-specific small RNAs into caput-derived embryos not only completely rescued preimplantation molecular defects but also suppressed the post-implantation embryonic lethality phenotype. These findings reveal an essential role for small RNA remodeling during post-testicular maturation of mammalian sperm and identify a specific preimplantation gene expression program responsive to sperm-delivered microRNAs.
Collapse
Affiliation(s)
- Colin C Conine
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Fengyun Sun
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lina Song
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jaime A Rivera-Pérez
- Department of Pediatrics, Division of Genes and Development, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Oliver J Rando
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
32
|
Park YJ, Battistone MA, Kim B, Breton S. Relative contribution of clear cells and principal cells to luminal pH in the mouse epididymis. Biol Reprod 2018; 96:366-375. [PMID: 28203710 DOI: 10.1095/biolreprod.116.144857] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/06/2016] [Accepted: 01/04/2017] [Indexed: 12/27/2022] Open
Abstract
While spermatozoa undergo epididymal maturation, they remain quiescent thanks to the establishment of a low luminal pH. This study is aimed at determining how epithelial cells lining the epididymal lumen work together to maintain and regulate this acidic milieu. In particular, we examined the relative contribution of clear cells (CCs) and principal cells (PCs) to this process. Functional analysis in the mouse cauda epididymidis (Cd) perfused in vivo showed that the pH of a control solution remained constant at pH 6.6 after perfusion through the Cd lumen. In contrast, the pH of both an acidic (pH 5.8) and alkaline (pH 7.8) perfusate was progressively restored toward the control acidic pH. Pharmacological studies indicated the contribution of cystic fibrosis transmembrane regulator, previously shown to be present in the apical membrane of PCs, to the recovery from an acidic pH of 5.8. In addition, we found that CCs and PCs equally contribute to the recovery from an alkaline of 7.8, via the H+ pumping vacuolar ATPase (V-ATPase) located in CCs, and the Na+/H+ exchanger type 3 (NHE3) located in PCs. Immunofluorescence labeling showed apical membrane accumulation of the V-ATPase in CCs at pH 7.8, and its internalization at pH 5.8 compared to pH 6.6. Immunofluorescence showed expression of NHE3, but absence of NHE2, in PCs located in the Cd. RT-PCR and western blotting showed expression of NHE3 in all epididymal regions. Luminal 8-(4-chlorophenylthio)adenosine 3΄,5΄-cyclic monophosphate (cpt-cAMP) partially inhibited luminal pH recovery from pH 7.8. However, cpt-cAMP induced an increase in V-ATPase apical membrane accumulation at this pH. Cell fractionation studies showed the apical accumulation of NHE3 from intracellular vesicles at pH 7.8 versus 6.6, and prevention of this effect by cpt-cAMP. These results indicate the participation of both CCs and PCs in the regulation of luminal pH in the epididymis. Our study also shows the dual role of PCs in HCO3− and H+ secretion, and that this switch from base to acid secretion depends on the luminal environment. Characterization of the respective roles of CCs and PCs in the regulation of the optimal luminal condition for epididymal sperm maturation should provide new frameworks for the evaluation and treatment of male infertility.
Collapse
Affiliation(s)
- Yoo-Jin Park
- Department of Laboratory Medicine,College of Medicine,The Catholic University of Korea,Seoul,Republic of Korea
| | - Maria Agustina Battistone
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Bongki Kim
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea.,Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, Korea
| | - Sylvie Breton
- Lesieur, R&D Center ESPCI ParisTech - CNRS, Coudekerque-Branche, France
| |
Collapse
|
33
|
Ritagliati C, Baro Graf C, Stival C, Krapf D. Regulation mechanisms and implications of sperm membrane hyperpolarization. Mech Dev 2018; 154:33-43. [PMID: 29694849 DOI: 10.1016/j.mod.2018.04.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022]
Abstract
Mammalian sperm are unable to fertilize the egg immediately after ejaculation. In order to gain fertilization competence, they need to undergo a series of biochemical and physiological modifications inside the female reproductive tract, known as capacitation. Capacitation correlates with two essential events for fertilization: hyperactivation, an asymmetric and vigorous flagellar motility, and the ability to undergo the acrosome reaction. At a molecular level, capacitation is associated to: phosphorylation cascades, modification of membrane lipids, alkalinization of the intracellular pH, increase in the intracellular Ca2+ concentration and hyperpolarization of the sperm plasma membrane potential. Hyperpolarization is a crucial event in capacitation since it primes the sperm to undergo the exocytosis of the acrosome content, essential to achieve fertilization of the oocyte.
Collapse
Affiliation(s)
- Carla Ritagliati
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET-UNR), Rosario 2000, Argentina.
| | - Carolina Baro Graf
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET-UNR), Rosario 2000, Argentina
| | - Cintia Stival
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET-UNR), Rosario 2000, Argentina
| | - Dario Krapf
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET-UNR), Rosario 2000, Argentina; Laboratorio de Especialidades Reproductivas, Facultad de Ciencias Bioquimicas y Farmacéuticas, UNR, Rosario 2000, Argentina.
| |
Collapse
|
34
|
Jin SK, Yang WX. Factors and pathways involved in capacitation: how are they regulated? Oncotarget 2018; 8:3600-3627. [PMID: 27690295 PMCID: PMC5356907 DOI: 10.18632/oncotarget.12274] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/23/2016] [Indexed: 01/07/2023] Open
Abstract
In mammals, fertilization occurs via a comprehensive progression of events. Freshly ejaculated sperm have yet to acquire progressive motility or fertilization ability. They must first undergo a series of biochemical and physiological changes, collectively known as capacitation. Capacitation is a significant prerequisite to fertilization. During the process of capacitation, changes in membrane properties, intracellular ion concentration and the activities of enzymes, together with other protein modifications, induce multiple signaling events and pathways in defined media in vitro or in the female reproductive tract in vivo. These, in turn, stimulate the acrosome reaction and prepare spermatozoa for penetration of the egg zona pellucida prior to fertilization. In the present review, we conclude all mainstream factors and pathways regulate capacitation and highlight their crosstalk. We also summarize the relationship between capacitation and assisted reproductive technology or human disease. In the end, we sum up the open questions and future avenues in this field.
Collapse
Affiliation(s)
- Shi-Kai Jin
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
35
|
Battistone MA, Nair AV, Barton CR, Liberman RN, Peralta MA, Capen DE, Brown D, Breton S. Extracellular Adenosine Stimulates Vacuolar ATPase-Dependent Proton Secretion in Medullary Intercalated Cells. J Am Soc Nephrol 2017; 29:545-556. [PMID: 29222395 DOI: 10.1681/asn.2017060643] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/18/2017] [Indexed: 12/29/2022] Open
Abstract
Acidosis is an important complication of AKI and CKD. Renal intercalated cells (ICs) express the proton pumping vacuolar H+-ATPase (V-ATPase) and are extensively involved in acid-base homeostasis. H+ secretion in type A intercalated cells (A-ICs) is regulated by apical vesicle recycling and stimulated by cAMP. In other cell types, cAMP is increased by extracellular agonists, including adenosine, through purinergic receptors. Adenosine is a Food and Drug Administration-approved drug, but very little is known about the effect of adenosine on IC function. Therefore, we investigated the role of adenosine in the regulation of V-ATPase in ICs. Intravenous treatment of mice with adenosine or agonists of ADORA2A and ADORA2B purinergic P1 receptors induced V-ATPase apical membrane accumulation in medullary A-ICs but not in cortical A-ICs or other IC subtypes. Both receptors are located in A-IC apical membranes, and adenosine injection increased urine adenosine concentration and decreased urine pH. Cell fractionation showed that adenosine or an ADORA2A or ADORA2B agonist induced V-ATPase translocation from vesicles to the plasma membrane and increased protein kinase A (PKA)-dependent protein phosphorylation in purified medullary ICs that were isolated from mice. Either ADORA2A or ADORA2B antagonists or the PKA inhibitor mPKI blocked these effects. Finally, a fluorescence pH assay showed that adenosine activates V-ATPase in isolated medullary ICs. Our study shows that medullary A-ICs respond to luminal adenosine through ADORA2A and ADORA2B receptors in a cAMP/PKA pathway-dependent mechanism to induce V-ATPase-dependent H+ secretion.
Collapse
Affiliation(s)
- Maria A Battistone
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anil V Nair
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Claire R Barton
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rachel N Liberman
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maria A Peralta
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Diane E Capen
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dennis Brown
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sylvie Breton
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
36
|
Gervasi MG, Visconti PE. Molecular changes and signaling events occurring in spermatozoa during epididymal maturation. Andrology 2017; 5:204-218. [PMID: 28297559 DOI: 10.1111/andr.12320] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/01/2016] [Accepted: 11/30/2016] [Indexed: 12/11/2022]
Abstract
After leaving the testis, spermatozoa have not yet acquired the ability to move progressively and are unable to fertilize oocytes. To become fertilization competent, they must go through an epididymal maturation process in the male, and capacitation in the female tract. Epididymal maturation can be defined as those changes occurring to spermatozoa in the epididymis that render the spermatozoa the ability to capacitate in the female tract. As part of this process, sperm cells undergo a series of biochemical and physiological changes that require incorporation of new molecules derived from the epididymal epithelium, as well as post-translational modifications of endogenous proteins synthesized during spermiogenesis in the testis. This review will focus on epididymal maturation events, with emphasis in recent advances in the understanding of the molecular basis of this process.
Collapse
Affiliation(s)
- M G Gervasi
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - P E Visconti
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
37
|
Impaired sperm maturation in conditional Lcn6 knockout mice†. Biol Reprod 2017; 98:28-41. [DOI: 10.1093/biolre/iox128] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 10/12/2017] [Indexed: 12/23/2022] Open
|
38
|
Posttranslational Modifications in Spermatozoa and Effects on Male Fertility and Sperm Viability. ACTA ACUST UNITED AC 2017; 21:245-256. [DOI: 10.1089/omi.2016.0173] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
39
|
Singh DK, Deshmukh RK, Narayanan PK, Shivaji S, Siva AB. SRC family kinases in hamster spermatozoa: evidence for the presence of LCK. Reproduction 2017; 153:655-669. [PMID: 28250239 DOI: 10.1530/rep-16-0591] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/03/2017] [Accepted: 02/28/2017] [Indexed: 01/16/2023]
Abstract
Sperm capacitation is a prerequisite for successful fertilization. Increase in tyrosine phosphorylation is considered the hallmark of capacitation and attempts to understand its regulation are ongoing. In this regard, we attempted to study the role of SRC family kinases (SFKs) in the hamster sperm functions. Interestingly, we found the presence of the lymphocyte-specific protein tyrosine kinase, LCK, in mammalian spermatozoa and further characterized it in terms of its localization and function. LCK was found in spermatozoa of several species, and its transcript was identified in the hamster testis. Autophosphorylation of LCK at the Y394 residue increased as capacitation progressed, indicating an upregulation of LCK activity during capacitation. Inhibition of LCK (and perhaps the other SFKs) with the use of a specific inhibitor showed a significant decrease in protein tyrosine phosphorylation of several proteins, implying LCK/SFKs as key tyrosine kinase(s) regulating tyrosine phosphorylation during hamster sperm capacitation. Dihydrolipoamide dehydrogenase was identified as a substrate for LCK/SFK. LCK/SFKs inhibition significantly reduced the percentage fertilization (in vitro) but had no effect on sperm motility, hyperactivation and acrosome reaction. In summary, this is the first report on the presence of LCK, an SFK of hematopoietic lineage in spermatozoa besides being the first study on the role of SFKs in the spermatozoa of Syrian hamsters.
Collapse
Affiliation(s)
| | | | | | - Sisinthy Shivaji
- CSIR-Centre for Cellular and Molecular BiologyHyderabad 500007, India
| | | |
Collapse
|
40
|
Jerczynski O, Lacroix-Pépin N, Boilard E, Calvo E, Bernet A, Fortier MA, Björkgren I, Sipilä P, Belleannée C. Role of Dicer1-Dependent Factors in the Paracrine Regulation of Epididymal Gene Expression. PLoS One 2016; 11:e0163876. [PMID: 27695046 PMCID: PMC5047620 DOI: 10.1371/journal.pone.0163876] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 09/15/2016] [Indexed: 12/21/2022] Open
Abstract
Dicer1 is an endoribonuclease involved in the biogenesis of functional molecules such as microRNAs (miRNAs) and endogenous small interfering RNAs (endo-siRNAs). These small non-coding RNAs are important regulators of post-transcriptional gene expression and participate in the control of male fertility. With the knowledge that 1) Dicer1-dependent factors are required for proper sperm maturation in the epididymis, and that 2) miRNAs are potent mediators of intercellular communication in most biological systems, we investigated the role of Dicer1-dependent factors produced by the proximal epididymis (initial segment/caput)- including miRNAs- on the regulation of epididymal gene expression in the distal epididymis regions (i.e. corpus and cauda). To this end, we performed comparative microarray and ANOVA analyses on control vs. Defb41iCre/wt;Dicer1fl/fl mice in which functional Dicer1 is absent from the principal cells of the proximal epididymis. We identified 35 and 33 transcripts that displayed significant expression level changes in the corpus and cauda regions (Fold change > 2 or < -2; p < 0.002), respectively. Among these transcripts, Zn-alpha 2-glycoprotein (Azgp1) encodes for a sperm equatorial protein whose expression in the epididymis of Dicer1 cKO mice is significantly increased compared to controls. In addition, 154 miRNAs, including miR-210, miR-672, miR-191 and miR-204, showed significantly impaired biogenesis in the absence of Dicer1 from the principal cells of the proximal epididymis (Fold change > 2 or < -2; p < 0.01). These miRNAs are secreted via extracellular vesicles (EVs) derived from the DC2 epididymal principal cell line, and their expression correlates with target transcripts involved in distinct biological pathways, as evidenced by in silico analysis. Albeit correlative and based on in silico approach, our study proposes that Dicer1-dependent factors trigger- directly or not-significant genes expression changes in distinct regions of this organ. The paracrine control of functions important to post-testicular sperm maturation by Dicer1-dependent factors may open new avenues for the identification of molecular targets important to male fertility control.
Collapse
Affiliation(s)
- Olivia Jerczynski
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, CHU de Québec Research Center (CHUL), Quebec City, Quebec, Canada
| | - Nicolas Lacroix-Pépin
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, CHU de Québec Research Center (CHUL), Quebec City, Quebec, Canada
| | - Eric Boilard
- Department of Immunity and Infectious Diseases, Université Laval, CHU de Québec Research Center (CHUL), Quebec City, Quebec, Canada
| | - Ezequiel Calvo
- Endocrinology unit, CHU de Québec Research Center (CHUL), Quebec City, Quebec, Canada
| | - Agathe Bernet
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, CHU de Québec Research Center (CHUL), Quebec City, Quebec, Canada
| | - Michel A. Fortier
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, CHU de Québec Research Center (CHUL), Quebec City, Quebec, Canada
| | - Ida Björkgren
- Department of Physiology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Petra Sipilä
- Department of Physiology, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Clémence Belleannée
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, CHU de Québec Research Center (CHUL), Quebec City, Quebec, Canada
| |
Collapse
|
41
|
Sullivan R, Mieusset R. The human epididymis: its function in sperm maturation. Hum Reprod Update 2016; 22:574-87. [PMID: 27307387 DOI: 10.1093/humupd/dmw015] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 04/25/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Spermatozoa acquire their fertilizing ability and forward motility properties during epididymal transit. Our knowledge of gamete physiology is based on studies conducted in laboratory and domestic species; our knowledge of these processes in humans is limited. Medical indications for assisted reproductive technologies (ART) have progressed to include male infertility. Surgical procedures allow collection of spermatozoa from all along the human excurrent ducts, and the former have been used with some success in reproductive medicine. This has raised questions over the role of the epididymis in human sperm physiology. OBJECTIVE AND RATIONALE To reanalyze what we now know about epididymal physiology in humans and to assess the relevance of laboratory animal models for understanding human physiology and the pathophysiology of the epididymis. SEARCH METHODS A systematic bibliographic search of PubMed for articles published in English before May 2015 was carried out using the search terms 'epididymis' and 'sperm maturation'. Literature on the consequences of vasectomy on the epididymis was also searched. OUTCOMES Whereas the proximal epididymis is almost exclusively occupied by efferent ducts, the sperm reservoir capacity is poorly developed in humans. At the molecular level, the human transcriptome and proteome show some segment specificity; conflicting results persist with regard to secretome variation along the tubule. The number of genes regulated along the excurrent ducts in men is lower when compared to rodent species, but remains significant. It is challenging to reconcile biochemical and physiological studies with clinical data obtained from men undergoing reanastomosis of the vas deferens at different points along the excurrent duct. We propose that vasectomy/vasovasostomy is a model to understand the consequences of obstruction on epididymis function in humans. WIDER IMPLICATIONS Despite the scarcity of biological material available, the interspecies variability of the male reproductive tract urges us to use modern molecular and cellular biology tools to better understand human epididymis physiology in order to apply ART in a more responsible manner.
Collapse
Affiliation(s)
- Robert Sullivan
- Département d'obstétrique, gynécologie et reproduction, Centre de recherche du Centre hospitalier de l'Université Laval, axe reproduction, santé de la mère et de l'enfant,
| | - Roger Mieusset
- Médecine de la Reproduction, CHU Toulouse, 31059 Toulouse, France Groupe de Recherche en Fertilité Humaine EA 3694, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
42
|
Xu B, Washington AM, Hinton BT. Initial Segment Differentiation Begins During a Critical Window and Is Dependent upon Lumicrine Factors and SRC Proto-Oncogene (SRC) in the Mouse. Biol Reprod 2016; 95:15. [PMID: 27281706 PMCID: PMC5029432 DOI: 10.1095/biolreprod.116.138388] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/20/2016] [Indexed: 11/13/2022] Open
Abstract
Without a fully developed and functioning initial segment, the most proximal region of the epididymis, male infertility results. Therefore, it is important to understand the development of the initial segment. During postnatal development of the epididymis, many cellular processes of the initial segment are regulated by lumicrine factors, which are produced by the testis and enter the epididymis with testicular luminal fluid. In this report, we showed that prior to Postnatal Day 15 (P15), the initial segment was lumicrine factor independent in the mouse. However, from P19 onward, lumicrine factors were essential for the proliferation and survival of initial segment epithelial cells. Therefore, P15 to P19 was a critical window that established the dependency of lumicrine factors in the initial segment epithelium. The initial segment-specific kinase activity profile, a marker of initial segment differentiation, was also established during this window. The SFK (SRC proto-oncogene family kinases), ERK pathway (known as the RAF/MEK/ERK pathway) components, and AMPK (AMP-activated protein kinases) pathway components had increased activities from P15 to P19, suggesting that lumicrine factors regulated SFK/ERK/AMPK signaling to initiate differentiation of the initial segment from P15 to P19. Compared with litter mate controls, juvenile Src null mice displayed lower levels of MAPK3/1 (mitogen-activated protein kinase 3/1) activity and a reduced level of differentiation in the initial segment epithelium, a similar phenotype resulting from inhibition of SRC activity within the window of P15 to P19. Therefore, lumicrine factor-dependent SRC activity signaling through MAPK3/1 is important for the initiation of initial segment differentiation during a critical window of development.
Collapse
Affiliation(s)
- Bingfang Xu
- Department of Cell Biology, University of Virginia Health System, Charlottesville, Virginia
| | - Angela M Washington
- Department of Cell Biology, University of Virginia Health System, Charlottesville, Virginia
| | - Barry T Hinton
- Department of Cell Biology, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|
43
|
Belleannée C. Extracellular microRNAs from the epididymis as potential mediators of cell-to-cell communication. Asian J Androl 2016; 17:730-6. [PMID: 26178395 PMCID: PMC4577581 DOI: 10.4103/1008-682x.155532] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Ribonucleic acid (RNA) was previously thought to remain inside cells as an intermediate between genes and proteins during translation. However, it is now estimated that 98% of the mammalian genomic output is transcribed as noncoding RNAs, which are involved in diverse gene expression regulatory mechanisms and can be transferred from one cell to another through extracellular communication. For instance, microRNAs are 22-nucleotide-long noncoding RNAs that are generated by endonuclease cleavage of precursors inside the cells and are secreted as extracellular microRNAs to regulate target cell posttranscriptional gene expression via RNA interference. We and others have shown that different populations of microRNAs are expressed in distinct regions of the human epididymis and regulate the expression of target genes that are involved in the control of male fertility as indicated by knock-out mouse models. Importantly, some microRNAs, including the microRNA-888 (miR-888) cluster that is exclusively expressed in the reproductive system of human and nonhuman primates, are released in the sperm-surrounding fluid in the epididymis via extracellular vesicles, the so-called epididymosomes. In addition to interacting with the membrane of maturing spermatozoa, these extracellular vesicles containing microRNAs communicate with epithelial cells located downstream from their release site, suggesting a role in the luminal exocrine control of epididymal functions. Apart from their potential roles as mediators of intercellular communication within the epididymis, these extracellular microRNAs are potent molecular targets for the noninvasive diagnosis of male infertility.
Collapse
Affiliation(s)
- Clémence Belleannée
- Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Reproduction, Mother and Youth Health Division, Research Center CHU de Québec, Québec, G1V 4G2, Canada
| |
Collapse
|
44
|
Tyrosine kinase-mediated axial motility of basal cells revealed by intravital imaging. Nat Commun 2016; 7:10666. [PMID: 26868824 PMCID: PMC4754344 DOI: 10.1038/ncomms10666] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/08/2016] [Indexed: 12/28/2022] Open
Abstract
Epithelial cells are generally considered to be static relative to their neighbours. Basal cells in pseudostratified epithelia display a single long cytoplasmic process that can cross the tight junction barrier to reach the lumen. Using in vivo microscopy to visualize the epididymis, a model system for the study of pseudostratified epithelia, we report here the surprising discovery that these basal cell projections--which we call axiopodia--periodically extend and retract over time. We found that axiopodia extensions and retractions follow an oscillatory pattern. This movement, which we refer to as periodic axial motility (PAM), is controlled by c-Src and MEK1/2-ERK1/2. Therapeutic inhibition of tyrosine kinase activity induces a retraction of these projections. Such unexpected cell motility may reflect a novel mechanism by which specialized epithelial cells sample the luminal environment.
Collapse
|
45
|
Li J, Mao R, Zhou Q, Ding L, Tao J, Ran MM, Gao ES, Yuan W, Wang JT, Hou LF. Exposure to bisphenol A (BPA) in Wistar rats reduces sperm quality with disruption of ERK signal pathway. Toxicol Mech Methods 2016; 26:180-8. [DOI: 10.3109/15376516.2016.1139024] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
46
|
Sperm Capacitation and Acrosome Reaction in Mammalian Sperm. SPERM ACROSOME BIOGENESIS AND FUNCTION DURING FERTILIZATION 2016; 220:93-106. [DOI: 10.1007/978-3-319-30567-7_5] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
47
|
Sharma U, Conine CC, Shea JM, Boskovic A, Derr AG, Bing XY, Belleannee C, Kucukural A, Serra RW, Sun F, Song L, Carone BR, Ricci EP, Li XZ, Fauquier L, Moore MJ, Sullivan R, Mello CC, Garber M, Rando OJ. Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals. Science 2015; 351:391-396. [PMID: 26721685 DOI: 10.1126/science.aad6780] [Citation(s) in RCA: 862] [Impact Index Per Article: 86.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 12/10/2015] [Indexed: 12/13/2022]
Abstract
Several recent studies link parental environments to phenotypes in subsequent generations. In this work, we investigate the mechanism by which paternal diet affects offspring metabolism. Protein restriction in mice affects small RNA (sRNA) levels in mature sperm, with decreased let-7 levels and increased amounts of 5' fragments of glycine transfer RNAs (tRNAs). In testicular sperm, tRNA fragments are scarce but increase in abundance as sperm mature in the epididymis. Epididymosomes (vesicles that fuse with sperm during epididymal transit) carry RNA payloads matching those of mature sperm and can deliver RNAs to immature sperm in vitro. Functionally, tRNA-glycine-GCC fragments repress genes associated with the endogenous retroelement MERVL, in both embryonic stem cells and embryos. Our results shed light on sRNA biogenesis and its dietary regulation during posttesticular sperm maturation, and they also link tRNA fragments to regulation of endogenous retroelements active in the preimplantation embryo.
Collapse
Affiliation(s)
- Upasna Sharma
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Colin C Conine
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jeremy M Shea
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ana Boskovic
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Alan G Derr
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Xin Y Bing
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Clemence Belleannee
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, Centre Hospitalier Universitaire de Québec Research Center,,Quebec City, Canada, G1V 4G2
| | - Alper Kucukural
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ryan W Serra
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Fengyun Sun
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lina Song
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Benjamin R Carone
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Emiliano P Ricci
- RNAi Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Xin Z Li
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA.,RNAi Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lucas Fauquier
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Melissa J Moore
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA.,RNAi Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Robert Sullivan
- Department of Obstetrics, Gynecology and Reproduction, Université Laval, Centre Hospitalier Universitaire de Québec Research Center,,Quebec City, Canada, G1V 4G2
| | - Craig C Mello
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.,RNAi Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Manuel Garber
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Oliver J Rando
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
48
|
Baker MA. Proteomics of post-translational modifications of mammalian spermatozoa. Cell Tissue Res 2015; 363:279-287. [DOI: 10.1007/s00441-015-2249-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 06/18/2015] [Indexed: 12/25/2022]
|
49
|
Vasen G, Battistone MA, Croci DO, Brukman NG, Weigel Muñoz M, Stupirski JC, Rabinovich GA, Cuasnicú PS. The galectin-1-glycan axis controls sperm fertilizing capacity by regulating sperm motility and membrane hyperpolarization. FASEB J 2015; 29:4189-200. [PMID: 26136479 DOI: 10.1096/fj.15-270975] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/15/2015] [Indexed: 12/15/2022]
Abstract
Lectin-glycan recognition systems play central roles in many physiologic and pathologic processes. We identified a role for galectin-1 (Gal-1), a highly conserved glycan-binding protein, in the control of sperm function. We found that Gal-1 is expressed in the epididymis and associates with sperm during epididymal maturation. Exposure of sperm to Gal-1 resulted in glycan-dependent modulation of the acrosome reaction (AR), a key event in the fertilization process. Gal-1-deficient (Lgals1(-/-)) mice revealed the essential contribution of this lectin for full sperm fertilizing ability both in vitro and in vivo. Mechanistically, Lgals1(-/-) sperm exhibited defects in their ability to develop hyperactivation, a vigorous motility required for penetration of the egg vestments. Moreover, Lgals1(-/-) sperm showed a decreased ability to control cell volume and to undergo progesterone-induced AR, phenotypes that were rescued by exposure of the cells to recombinant Gal-1. Interestingly, the AR defect was associated with a deficiency in sperm membrane potential hyperpolarization. Our study highlights the relevance of the Gal-1-glycan axis in sperm function with critical implications in mammalian reproductive biology.
Collapse
Affiliation(s)
- Gustavo Vasen
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria Agustina Battistone
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Diego O Croci
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nicolás G Brukman
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariana Weigel Muñoz
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan C Stupirski
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Patricia S Cuasnicú
- *Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
50
|
Barkalina N, Jones C, Wood MJA, Coward K. Extracellular vesicle-mediated delivery of molecular compounds into gametes and embryos: learning from nature. Hum Reprod Update 2015; 21:627-39. [PMID: 26071427 DOI: 10.1093/humupd/dmv027] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/21/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Currently, even the most sophisticated methods of assisted reproductive technology (ART) allow us to achieve live births in only approximately 30% of patients, indicating that our understanding of the fine mechanisms underlying reproduction is far from ideal. One of the main challenges associated with studies of gamete structure and function is that these cells are remarkably resistant towards the uptake of exogenous substances, including 'molecular research tools' such as drugs, biomolecules and intracellular markers. This phenomenon can affect not only the performance of reproductive biology research techniques, but also the outcomes of the in vitro handling of gametes, which forms the cornerstone of ART. Improvement of intra-gamete delivery in a non-aggressive fashion is vital for the investigation of gamete physiology, and the advancement of infertility treatment. In this review, we outline the current state of nanomaterial-mediated delivery into gametes and embryos in vitro, and discuss the potential of a novel exciting drug delivery technology, based upon the use of targeted 'natural' nanoparticles known as extracellular vesicles (EVs), for reproductive science and ART, given the promising emerging data from other fields. METHODS A comprehensive electronic search of PubMed and Web of Science databases was performed using the following keywords: 'nanoparticles', 'nanomaterials', 'cell-penetrating peptides', 'sperm', 'oocyte', 'egg', 'embryo', 'exosomes', 'microvesicles', 'extracellular vesicles', 'delivery', 'reproduction', to identify the relevant research and review articles, published in English up to January 2015. The reference lists of identified publication were then scanned to extract additional relevant publications. RESULTS Biocompatible engineered nanomaterials with high loading capacity, stability and selective affinity represent a potential versatile tool for the minimally invasive internalization of molecular cargo into gametes and embryos. However, it is becoming increasingly clear that the translation of these experimental tools into clinical applications is likely to be limited by their non-biodegradable nature. To allow the subsequent use of these methodologies for clinical ART, studies should utilize biodegradable delivery platforms, which mimic natural mechanisms of molecular cargo trafficking as closely as possible. Currently, EVs represent the most physiological intracellular delivery tools for reproductive science and medicine. These natural mediators of cell communication combine the benefits of engineered nanomaterials, such as the potential for in vitro production, targeting and loading, with the essential feature of biodegradability. CONCLUSION We anticipate that future investigations into the possibility of applying EVs for the intentional intracellular delivery of molecular compounds into gametes and embryos will open new horizons for reproductive science and clinical ART, ultimately leading to improvements in patient care.
Collapse
Affiliation(s)
- Natalia Barkalina
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Celine Jones
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Le Gros Clark Building, South Parks Road, Oxford OX1 3QX, UK
| | - Kevin Coward
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| |
Collapse
|