1
|
Mukherjee A, Verma A, Das T, Ghosh B, Ghosh Z. Circulating microRNAs in Body Fluid: "Fingerprint" RNA Snippets Deeply Impact Reproductive Biology. Reprod Sci 2025; 32:555-574. [PMID: 39658771 DOI: 10.1007/s43032-024-01753-y] [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: 07/01/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024]
Abstract
Circulating miRNAs (C-miRNAs) occuring in a cell-free form within body fluids and other extracellular environments have garnered attention in recent times. They offer deeper insight into various physiological and pathological processes which include reproductive health. This review delves into their diagnostic potential across a spectrum of reproductive disorders, including conditions affecting ovarian function, male infertility and post pregnancy issues. Through analysis of C-miRNA profiles in bodily fluids, researchers uncover crucial markers indicative of reproductive challenges. Dysregulated C-miRNAs emerge as important players in the progression of several reproductive disorders which is the main focus of this review. Advancements in technology, facilitate precise detection and quantification of C-miRNAs, paving the way for innovative diagnostic approaches. Challenges in studying C-miRNAs, such as their low abundance and variability in expression levels, underscore the need for standardized protocols and rigorous validation methods. Despite these challenges, ongoing research endeavors aim to unravel the complex regulatory roles of C-miRNAs in reproductive biology, with potential implications for clinical practice and therapeutic interventions.
Collapse
Affiliation(s)
- Ayan Mukherjee
- Department of Animal Biotechnology, West Bengal University of Animal and Fishery Sciences, Mohanpur, West Bengal, 741252, India.
| | - Arpana Verma
- Department of Biological Sciences, Bose Institute, EN Block, Sector V, Kolkata, West Bengal, 700091, India
| | - Troyee Das
- Department of Biological Sciences, Bose Institute, EN Block, Sector V, Kolkata, West Bengal, 700091, India
| | - Byapti Ghosh
- Department of Biological Sciences, Bose Institute, EN Block, Sector V, Kolkata, West Bengal, 700091, India
| | - Zhumur Ghosh
- Department of Biological Sciences, Bose Institute, EN Block, Sector V, Kolkata, West Bengal, 700091, India.
| |
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
|
Sethi M, Mohanty TK, Shah N, Bhakat M, Kumar N, Baithalu RK. Understanding the Crucial Role of Seminal Plasma Exosomes in Bull Fertility: A Review. Reprod Domest Anim 2024; 59:e70000. [PMID: 39714190 DOI: 10.1111/rda.70000] [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: 09/17/2024] [Revised: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024]
Abstract
Bull fertility is a multi-factorial trait and is affected by many factors, such as nutrition, genetics, and epigenetics. Superior quality male germplasm with high genetic merit helps to improve the livestock production trait. To achieve the target of livestock production, the availability of superior male germplasm is a great concern. In developing countries, there is a gap between the highly fertile frozen semen doses produced and the highly fertile frozen semen doses required. Improving the quality of existing low-fertile semen from high genetic merit bulls seems to play a promising role in filling this gap. Seminal exosomes are extracellular vesicles secreted by the epithelial cells of the testis, epididymis, and accessory sex glands such as the prostate gland. They contain a cargo of bioactive molecules such as proteins, nucleic acids and various metabolites. These molecules are transferred to the spermatozoa during its maturation and help in sperm capacitation, maturation, acrosome reaction, and fertilisation. Studies reveal that seminal exosomes help to improve the sperm functionality of low-quality sperm. Identification of the molecular profile of exosomes of bulls with proven fertility and their addition in an extender containing low-fertile semen may help to ameliorate the sperm quality of low-fertile semen, which may eventually aid in generating quantities of highly fertile ejaculates.
Collapse
Affiliation(s)
- Manisha Sethi
- Animal Reproduction, Gynaecology and Obstetrics, Artificial Breeding Research Centre (ABRC), ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Tushar Kumar Mohanty
- Animal Reproduction, Gynaecology and Obstetrics, Artificial Breeding Research Centre (ABRC), ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Nadeem Shah
- Sardar Vallabhbhai Patel University of Agriculture & Technology, Meerut, Uttar Pradesh, India
| | - Mukesh Bhakat
- Central Institute for Research on Goats, Makhdum, Uttar Pradesh, India
| | - Nishant Kumar
- Animal Reproduction, Gynaecology and Obstetrics, Artificial Breeding Research Centre (ABRC), ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Rubina Kumari Baithalu
- Animal Reproduction, Gynaecology and Obstetrics, Artificial Breeding Research Centre (ABRC), ICAR-National Dairy Research Institute, Karnal, Haryana, India
| |
Collapse
|
4
|
Jankovičová J, Michalková K, Sečová P, Horovská Ľ, Antalíková J. The extracellular vesicle tetraspanin CD63 journey from the testis through the epididymis to mature bull sperm. Sci Rep 2024; 14:29449. [PMID: 39604592 PMCID: PMC11603341 DOI: 10.1038/s41598-024-81021-w] [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: 07/12/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024] Open
Abstract
The important role of extracellular vesicles, which are considered key mediators of intercellular communication under physiological and pathological conditions, in various cellular processes, including those crucial for mammalian reproduction, has been increasingly studied. Tetraspanins, including CD63, are widely used as markers of extracellular vesicles, but they may also play a role in their biogenesis, cargo selection, cell targeting, and uptake. This study aimed to map the journey of the extracellular vesicle protein tetraspanin CD63 from the testis through the epididymis into mature bull sperm via an approach that included immunohistochemistry (immunofluorescence and immunoperoxidase staining), Western blot analysis, and immunoprecipitation analysis. We described the presence of CD63 in bull testicular and epididymal tissues, extracellular vesicles produced in these organs and spermatozoa during epididymal transit and after ejaculation. In addition, we revealed the nonuniform distribution of potential CD63 partners, such as CD9, integrin αV and syntenin-1, in the sperm head and tail and in extracellular vesicles. These findings contribute to understanding the complex mechanisms underlying sperm maturation and point to the possible involvement of tetraspanins and their associated partners, either as part of extracellular vesicles or sperm membranes, in these processes.
Collapse
Grants
- VEGA-2/0074/24 Scientific Grant Agency of the Ministry of Education, Science, Research and Sport of the Slovak Republic and the Slovak Academy of Sciences
- VEGA-2/0074/24 Scientific Grant Agency of the Ministry of Education, Science, Research and Sport of the Slovak Republic and the Slovak Academy of Sciences
- VEGA-2/0074/24 Scientific Grant Agency of the Ministry of Education, Science, Research and Sport of the Slovak Republic and the Slovak Academy of Sciences
- VEGA-2/0074/24 Scientific Grant Agency of the Ministry of Education, Science, Research and Sport of the Slovak Republic and the Slovak Academy of Sciences
- VEGA-2/0074/24 Scientific Grant Agency of the Ministry of Education, Science, Research and Sport of the Slovak Republic and the Slovak Academy of Sciences
- APVV-19-0111 Slovak Research and Development Agency
- APVV-19-0111 Slovak Research and Development Agency
- APVV-19-0111 Slovak Research and Development Agency
- APVV-19-0111 Slovak Research and Development Agency
- APVV-19-0111 Slovak Research and Development Agency
Collapse
Affiliation(s)
- Jana Jankovičová
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, 840 05, Slovak Republic.
| | - Katarína Michalková
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, 840 05, Slovak Republic
| | - Petra Sečová
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, 840 05, Slovak Republic
| | - Ľubica Horovská
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, 840 05, Slovak Republic
| | - Jana Antalíková
- Laboratory of Reproductive Physiology, Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, Bratislava, 840 05, Slovak Republic
| |
Collapse
|
5
|
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
|
6
|
Ali W, Deng K, Bian Y, Liu Z, Zou H. Spectacular role of epididymis and bio-active cargo of nano-scale exosome in sperm maturation: A review. Biomed Pharmacother 2023; 164:114889. [PMID: 37209627 DOI: 10.1016/j.biopha.2023.114889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/30/2023] [Accepted: 05/12/2023] [Indexed: 05/22/2023] Open
Abstract
The epididymis is responsible for post-testicular sperm maturation as it provides a favorable environment for spermatozoa to gain the ability for movement and fertilization. The recent evidence has shown that, the spermatozoa are vulnerable to dynamic variations driven by various cellular exposure mechanisms mediated by epididymosomes. Exosomes provide new insight into a mechanism of intercellular communication because they provide direct evidence for the transfer of several important bio-active cargo elements (proteins, lipid, DNA, mRNA, microRNA, circular RNA, long noncoding RNA) between epididymis and spermatozoa. In broad sense, proteomic analysis of exosomes from epididymis indicates number of proteins that are involved in sperm motility, acrosomal reaction, prevent pre-mature sperm capacitation and male infertility. Pinpointing, how reproductive disorders are associated with bio-active cargo elements of nano-scale exosome in the male reproductive tract. Therefore, the current review presents evidence regarding the distinctive characteristics and functions of nano-scale exosome in the male reproductive tract in both pathological and physiological developments, and argue that these vesicles serve as an important regulator of male reproduction, fertility, and disease susceptibility.
Collapse
Affiliation(s)
- Waseem Ali
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Kai Deng
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Yusheng Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, PR China.
| |
Collapse
|
7
|
Transfer of Galectin-3-Binding Protein via Epididymal Extracellular Vesicles Promotes Sperm Fertilizing Ability and Developmental Potential in the Domestic Cat Model. Int J Mol Sci 2023; 24:ijms24043077. [PMID: 36834494 PMCID: PMC9966717 DOI: 10.3390/ijms24043077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Key proteins transferred by epididymal extracellular vesicles (EVs) to the transiting sperm cells contribute to their centrosomal maturation and developmental potential. Although not reported in sperm cells yet, galectin-3-binding protein (LGALS3BP) is known to regulate centrosomal functions in somatic cells. Using the domestic cat model, the objectives of this study were to (1) detect the presence and characterize the transfer of LGALS3BP via EVs between the epididymis and the maturing sperm cells and (2) demonstrate the impact of LGALS3BP transfer on sperm fertilizing ability and developmental potential. Testicular tissues, epididymides, EVs, and spermatozoa were isolated from adult individuals. For the first time, this protein was detected in EVs secreted by the epididymal epithelium. The percentage of spermatozoa with LGALS3BP in the centrosome region increased as cells progressively incorporated EVs during the epididymal transit. When LGALS3BP was inhibited during in vitro fertilization with mature sperm cells, less fertilized oocytes and slower first cell cycles were observed. When the protein was inhibited in epididymal EVs prior to incubation with sperm cells, poor fertilization success further demonstrated the role of EVs in the transfer of LGALS3BP to the spermatozoa. The key roles of this protein could lead to new approaches to enhance or control fertility in clinical settings.
Collapse
|
8
|
Belardin LB, Antoniassi MP, Camargo M, Intasqui P, Bertolla RP. Separating the chaff from the wheat: antibody-based removal of DNA-fragmented sperm. Hum Reprod 2023; 38:204-215. [PMID: 36539256 DOI: 10.1093/humrep/deac260] [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: 05/11/2021] [Revised: 11/12/2022] [Indexed: 12/24/2022] Open
Abstract
STUDY QUESTION Is it possible to remove sperm with damaged DNA from a semen sample? SUMMARY ANSWER By using immunomagnetic cell sorting that targets the sperm head-bound epididymal sperm-binding protein 1 (ELSPBP1), it was possible to produce an ELSPBP1(-) sperm fraction characterized by consistently lower levels of sperm DNA fragmentation (SDF). WHAT IS KNOWN ALREADY In bovines, ELSPBP1 is bound to dead spermatozoa. Human ejaculates with high SDF have increased detected levels of sperm ELSPBP1 when compared to ejaculates with low native SDF. STUDY DESIGN, SIZE, DURATION We recruited 267 patients who were referred to the clinic for conjugal infertility. After applying exclusion criteria, such as fever within 90 days of the study, history of systemic diseases, alterations or surgical interventions to the genital tract and use of cigarette or drugs, a total of 133 patients were included. A total of 52 samples were used for the evaluation of sperm ELSPBP1 levels (Sub-study 1), 41 samples for determination of ELSPBP1 location in human sperm (Sub-study 2), and 40 samples for immunomagnetic cell sorting targeting ELSPBP1, to produce ELSPBP1(-) (without ELSPBP1) and ELSPBP1(+) (with ELSPBP1) fractions (Sub-study 3). Samples were collected between July 2016 and September 2019. PARTICIPANTS/MATERIALS, SETTING, METHODS In Sub-study 1, sperm ELSPBP1 levels were assessed by western blotting. For Sub-study 2, ELSPBP1 was localized in sperm by immunocytochemistry. Finally, for Sub-study 3, sperm were selected based on incubation of semen samples with antibody-coated magnetic microspheres targeting ELSPBP1. Two fractions were produced (with or without ELSPBP1), and these sub-populations were submitted to an alkaline Comet assay for determination of SDF. MAIN RESULTS AND THE ROLE OF CHANCE Men with high SDF presented higher sperm ELSPBP1 levels when compared to the control group (low SDF), while no difference between groups was observed in seminal plasma. ELSPBP1 was located in the head region of human sperm. The ELSPBP1(+) fractions presented high and variable levels of SDF, while their paired ELSPBP(-) fractions presented consistently low SDF. LIMITATIONS, REASONS FOR CAUTION This work did not validate the levels of ELSPBP1 in other functional alterations of sperm, such as acrosome integrity or mitochondrial activity. Moreover, this is still a pre-clinical study, intended to demonstrate proof-of-concept that ELSPBP1 selects sperm with low DNA fragmentation; further investigation is warranted to demonstrate safety for use in ART. Sperm fractions were not assessed for sperm vitality. A clinical trial is still necessary for these findings to be extrapolated to outcomes in ART. WIDER IMPLICATIONS OF THE FINDINGS Our findings demonstrate that ELSPBP1 is associated with sperm with higher levels of DNA fragmentation. The finding that the sperm membrane can reflect alterations in DNA integrity could give rise to a novel molecular method for sperm preparation prior to use of assisted reproductive procedures. Moreover, the detection of sperm-bound ELSPBP1 could serve as an indirect method for the determination of DNA fragmentation. STUDY FUNDING/COMPETING INTEREST(S) L.B.B. was a recipient of a Ph.D. scholarship from the Sao Paulo Research Foundation-FAPESP (process number 2016/05487-3). R.P.B. is a recipient of a Scientific Productivity scholarship from the Brazilian National Council for Scientific and Technological Development-CNPq (process number 306705/2017-6). The authors have no conflict of interest to disclose. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- L B Belardin
- Human Reproduction Section, Division of Urology, Department of Surgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - M P Antoniassi
- Human Reproduction Section, Division of Urology, Department of Surgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - M Camargo
- Human Reproduction Section, Division of Urology, Department of Surgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - P Intasqui
- Human Reproduction Section, Division of Urology, Department of Surgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - R P Bertolla
- Human Reproduction Section, Division of Urology, Department of Surgery, Universidade Federal de São Paulo, São Paulo, Brazil
- Hospital São Paulo, São Paulo, Brazil
| |
Collapse
|
9
|
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
|
10
|
Belardin LB, Brochu K, Légaré C, Battistone MA, Breton S. Purinergic signaling in the male reproductive tract. Front Endocrinol (Lausanne) 2022; 13:1049511. [PMID: 36419764 PMCID: PMC9676935 DOI: 10.3389/fendo.2022.1049511] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022] Open
Abstract
Purinergic receptors are ubiquitously expressed throughout the body and they participate in the autocrine and paracrine regulation of cell function during normal physiological and pathophysiological conditions. Extracellular nucleotides activate several types of plasma membrane purinergic receptors that form three distinct families: P1 receptors are activated by adenosine, P2X receptors are activated by ATP, and P2Y receptors are activated by nucleotides including ATP, ADP, UTP, UDP, and UDP-glucose. These specific pharmacological fingerprints and the distinct intracellular signaling pathways they trigger govern a large variety of cellular responses in an organ-specific manner. As such, purinergic signaling regulates several physiological cell functions, including cell proliferation, differentiation and death, smooth muscle contraction, vasodilatation, and transepithelial transport of water, solute, and protons, as well as pathological pathways such as inflammation. While purinergic signaling was first discovered more than 90 years ago, we are just starting to understand how deleterious signals mediated through purinergic receptors may be involved in male infertility. A large fraction of male infertility remains unexplained illustrating our poor understanding of male reproductive health. Purinergic signaling plays a variety of physiological and pathophysiological roles in the male reproductive system, but our knowledge in this context remains limited. This review focuses on the distribution of purinergic receptors in the testis, epididymis, and vas deferens, and their role in the establishment and maintenance of male fertility.
Collapse
Affiliation(s)
- Larissa Berloffa Belardin
- Centre Hospitalier Universitaire de Québec - Research Centre and Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Kéliane Brochu
- Centre Hospitalier Universitaire de Québec - Research Centre and Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Christine Légaré
- Centre Hospitalier Universitaire de Québec - Research Centre and Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Maria Agustina Battistone
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sylvie Breton
- Centre Hospitalier Universitaire de Québec - Research Centre and Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
11
|
Roca J, Rodriguez-Martinez H, Padilla L, Lucas X, Barranco I. Extracellular vesicles in seminal fluid and effects on male reproduction. An overview in farm animals and pets. Anim Reprod Sci 2022; 246:106853. [PMID: 34556398 DOI: 10.1016/j.anireprosci.2021.106853] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/09/2021] [Accepted: 09/11/2021] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs) are lipid bilayer nanovesicles released by most functional cells to body fluids, containing bioactive molecules, mainly proteins, lipids, and nucleic acids having actions at target cells. The EVs have essential functions in cell-to-cell communication by regulating different biological processes in target cells. Fluids from the male reproductive tract, including seminal plasma, contain many extracellular vesicles (sEVs), which have been evaluated to a lesser extent than those of other body fluids, particularly in farm animals and pets. Results from the few studies that have been conducted indicated epithelial cells of the testis, epididymis, ampulla of ductus deferens and many accessory sex glands release sEVs mainly via apocrine mechanisms. The sEVs are morphologically heterogeneous and bind to functional cells of the male reproductive tract, spermatozoa, and cells of the functional tissues of the female reproductive tract after mating or insemination. The sEVs encapsulate proteins and miRNAs that modulate sperm functions and male fertility. The sEVs, therefore, could be important as reproductive biomarkers in breeding sires. Many of the current findings regarding sEV functions, however, need experimental confirmation. Further studies are particularly needed to characterize both membranes and contents of sEVs, as well as the interaction between sEVs and target cells (spermatozoa and functional cells of the internal female reproductive tract). A priority for conducting these studies is development of methods that can be standardized and that are scalable, cost-effective and time-saving for isolation of different subtypes of EVs present in the entire population of sEVs.
Collapse
Affiliation(s)
- Jordi Roca
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, 30100 Murcia, Spain.
| | - Heriberto Rodriguez-Martinez
- Department of Biomedical & Clinical Sciences (BKV), BKH/Obstetrics & Gynaecology, Faculty of Medicine and Health Sciences, Linköping University, SE-58185 Linköping, Sweden
| | - Lorena Padilla
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, 30100 Murcia, Spain
| | - Xiomara Lucas
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, 30100 Murcia, Spain
| | - Isabel Barranco
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, IT-40064 Bologna, Italy
| |
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
|
Griffin RA, Swegen A, Baker MA, Ogle RA, Smith N, Aitken RJ, Skerrett-Byrne DA, Fair S, Gibb Z. Proteomic analysis of spermatozoa reveals caseins play a pivotal role in preventing short-term periods of subfertility in stallions. Biol Reprod 2022; 106:741-755. [DOI: 10.1093/biolre/ioab225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/29/2021] [Accepted: 09/23/2021] [Indexed: 11/14/2022] Open
Abstract
Abstract
Stallions experience transient fluctuations in fertility throughout the breeding season. Considering pregnancy diagnoses cannot be ascertained until ~14 days post-breeding, the timely detection of decreases in stallion fertility would enhance industry economic and welfare outcomes. Therefore, this study aimed to identify the proteomic signatures reflective of short-term fertility fluctuations, and to determine the biological mechanisms governing such differences. Using LC–MS/MS, we compared the proteomic profile of semen samples collected from commercially “fertile” stallions, during high- and low-fertility periods. A total of 1702 proteins were identified, of which, 38 showed a significant change in abundance (p ≤ 0.05). Assessment of intra- and inter-stallion variability revealed that caseins (namely κ-, α-S1-, and α-S2-casein), were significantly more abundant during “high-fertility” periods, while several epididymal, and seminal plasma proteins (chiefly, epididymal sperm binding protein 1 [ELSPbP1], horse seminal plasma protein 1 [HSP-1] and clusterin), were significantly more abundant during “low-fertility” periods. We hypothesised that an increased abundance of caseins offers greater protection from potentially harmful seminal plasma proteins, thereby preserving cell functionality and fertility. In vitro exposure of spermatozoa to casein resulted in decreased levels of lipid scrambling (Merocyanine 540), higher abundance of sperm-bound caseins (α-S1-, α-S2-, and κ-casein), and lower abundance of sperm-bound HSP-1 (p ≤ 0.05). This study demonstrates key pathways governing short-term fertility fluctuations in the stallion, thereby providing a platform to develop robust, fertility assessment strategies into the future.
Collapse
Affiliation(s)
- Róisín Ann Griffin
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
| | - Aleona Swegen
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Mark A Baker
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
| | - Rachel Ann Ogle
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
| | - Nathan Smith
- Analytical and Biomedical Research Facility, Research Division, University of Newcastle, Callaghan, New South Wales, Australia
| | - Robert John Aitken
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
| | - David Anthony Skerrett-Byrne
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, New South Wales, Australia
| | - Sean Fair
- Laboratory of Animal Reproduction, Department of Biological Sciences, Biomaterials Research Cluster, Bernal Institute, University of Limerick, Limerick, Ireland
| | - Zamira Gibb
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
| |
Collapse
|
14
|
Chand S, Gowen A, Savine M, Moore D, Clark A, Huynh W, Wu N, Odegaard K, Weyrich L, Bevins RA, Fox HS, Pendyala G, Yelamanchili SV. A comprehensive study to delineate the role of an extracellular vesicle-associated microRNA-29a in chronic methamphetamine use disorder. J Extracell Vesicles 2021; 10:e12177. [PMID: 34913274 PMCID: PMC8674191 DOI: 10.1002/jev2.12177] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 10/27/2021] [Accepted: 11/25/2021] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs), which express a repertoire of cargo molecules (cf. proteins, microRNA, lipids, etc.), have been garnering a prominent role in the modulation of several cellular processes. Here, using both non-human primate and rodent model systems, we provide evidence that brain-derived EV (BDE) miRNA, miR-29a-3p (mir-29a), is significantly increased during chronic methamphetamine (MA) exposure. Further, miR-29a levels show significant increase both with drug-seeking and reinstatement in a rat MA self-administration model. We also show that EV-associated miR-29a is enriched in EV pool comprising of small EVs and exomeres and further plays a critical role in MA-induced inflammation and synaptodendritic damage. Furthermore, treatment with the anti-inflammatory drug ibudilast (AV411), which is known to reduce MA relapse, decreased the expression of miR-29a and subsequently attenuated inflammation and rescued synaptodendritic injury. Finally, using plasma from MUD subjects, we provide translational evidence that EV-miR29a could potentially serve as a biomarker to detect neuronal damage in humans diagnosed with MA use disorder (MUD). In summary, our work suggests that EV-associated miR-29a-3p plays a crucial role in MUD and might be used as a potential blood-based biomarker for detecting chronic inflammation and synaptic damage.
Collapse
Affiliation(s)
- Subhash Chand
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Austin Gowen
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Mason Savine
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Dalia Moore
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Alexander Clark
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Wendy Huynh
- Department of PsychologyUniversity of Nebraska–Lincoln (UNL)LincolnNebraskaUSA
| | - Niming Wu
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Katherine Odegaard
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | | | - Rick A. Bevins
- Department of PsychologyUniversity of Nebraska–Lincoln (UNL)LincolnNebraskaUSA
| | - Howard S. Fox
- Department of Neurological SciencesUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Gurudutt Pendyala
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Sowmya V. Yelamanchili
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| |
Collapse
|
15
|
Rowlison T, Comizzoli P. The Knowns and Unknowns about Epididymal Extracellular Vesicles in Different Animal Species. Adv Biol (Weinh) 2021; 6:e2101066. [PMID: 34816626 DOI: 10.1002/adbi.202101066] [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: 06/30/2021] [Revised: 10/04/2021] [Indexed: 11/07/2022]
Abstract
Sperm maturation during epididymal transit is a long and complex process. Although the roles of epididymal extracellular vesicles (EVs) on sperm quality have been extensively studied in recent years, there are still a lot of unexplored areas and too few species that are studied. The objective of this review is to focus on the contribution of epididymal EVs through the apocrine secretion of key factors, including proteins and small RNAs. Furthermore, the authors explore the alterations in the content of these vesicles related to male fertility and the effects of environmental stressors, and how these factors vary across taxa. Last, potential applications are covered, and the next steps in that field of research are highlighted.
Collapse
Affiliation(s)
- Tricia Rowlison
- Smithsonian Conservation Biology Institute, National Zoological Park, 3001 Connecticut Avenue NW, Washington, DC, 20008, USA
| | - Pierre Comizzoli
- Smithsonian Conservation Biology Institute, National Zoological Park, 3001 Connecticut Avenue NW, Washington, DC, 20008, USA
| |
Collapse
|
16
|
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
|
17
|
van der Horst G, Kotzè S, O'Riain MJ, Muller N, Maree L. A possible highway system for the rapid delivery of sperm from the testis to the penis in the naked mole-rat, Heterocephalus glaber. J Morphol 2021; 282:1478-1498. [PMID: 34296784 DOI: 10.1002/jmor.21399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/08/2021] [Accepted: 07/17/2021] [Indexed: 11/08/2022]
Abstract
Gametogenesis is suppressed in most members of the eusocial naked mole-rat (NMR) colony, while the queen selects mainly one breeding male during her life span. Recently, it was reported that the NMR testicular organization seems to produce spermatozoa on demand after suppression of spermatogenesis during most of gestation. A Sertoli cell "pump" is then used to flush the spermatozoa into short tubuli recti and simplified rete testis to reach the excurrent duct system. We hypothesize that the components of this duct system are adapted for rapid delivery of spermatozoa to the penis and for numerous copulations with the queen. Therefore, the aim was to study the ultrastructure of the male NMR reproductive duct system using light microscopy and transmission electron microscopy. The NMR rete testis gives rise to six to eight efferent tubules joining the caput epididymis. The caput epididymis resembles that of other rodents but with less distinction in terms of histological zoning. The remainder of the epididymis is considerably reduced in length compared to other rodents. In contrast, the vas deferens epithelium is highly specialized in that a vast range of vesicles, often closely associated with the spermatozoa, were visible. The large ampulla is a factory for merocrine and apocrine secretions, producing even more diverse vesicles. The transitional epithelial cells of the bladder appear to secrete abundant mucous and the penis as well as its baculum is relatively small. We speculate that these modifications strongly suggest that the excurrent duct system has been simplified and adjusted to compensate for the absence of long maturation and storage of spermatozoa. We propose that these adaptations to the NMR reproductive tract are associated with a state of degenerative orthogenesis that was selected for due to the absence of sperm competition and apparently rapid delivery of spermatozoa from the testis.
Collapse
Affiliation(s)
- Gerhard van der Horst
- Department of Medical, Biosciences, University of the Western Cape, Bellville, South Africa
| | - Sanet Kotzè
- Division of Clinical Anatomy, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa.,Ross University School of Veterinary Medicine, Basseterre, St. Kitts, West Indies
| | | | - Nolan Muller
- National Health Laboratory Services, Anatomical Pathology, Tygerberg Hospital, Parow, South Africa
| | - Liana Maree
- Department of Medical, Biosciences, University of the Western Cape, Bellville, South Africa
| |
Collapse
|
18
|
Paul N, Talluri TR, Nag P, Kumaresan A. Epididymosomes: A potential male fertility influencer. Andrologia 2021; 53:e14155. [PMID: 34213814 DOI: 10.1111/and.14155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 01/06/2023] Open
Abstract
During transit and storage in epididymis, spermatozoa undergo final maturation, acquire motility, functional competence and the ability to fertilise an oocyte. Epididymal secretions contain a complex biochemical milieu of diverse inorganic ions, proteins, metabolites and other molecules. Since it is believed that spermatozoa are translationally silent, proteins appearing in them are thought to be synthesised elsewhere, including epididymis, and then incorporated to the cells. One of the important mechanisms suggested to be involved in transfer of epididymal secretions to spermatozoa is through exosomes called epididymosomes. Epididymosomes released from the epididymal epithelium contain proteins, noncoding RNAs and distinct set of lipids that are transferred to spermatozoa while they pass through the different epididymal regions. Owing to the importance of these molecules for sperm maturation and fertilising ability, research on epididymosomes has gained increasing attention during the last decade. This review is focused on epididymosomes, with emphasis on recent advances in the understanding of mechanisms of epididymosomal cargo transfer to spermatozoa and potential roles of epididymosomes in sperm function and beyond. Possibilities of utilising the molecular signatures of epididymosomes as a tool for male fertility assessment are also discussed.
Collapse
Affiliation(s)
- Nilendu Paul
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Thirumala Rao Talluri
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Pradeep Nag
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| | - Arumugam Kumaresan
- Theriogenology Laboratory, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, India
| |
Collapse
|
19
|
Batra V, Bhushan V, Ali SA, Sarwalia P, Pal A, Karanwal S, Solanki S, Kumaresan A, Kumar R, Datta TK. Buffalo sperm surface proteome profiling reveals an intricate relationship between innate immunity and reproduction. BMC Genomics 2021; 22:480. [PMID: 34174811 PMCID: PMC8235841 DOI: 10.1186/s12864-021-07640-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/08/2021] [Indexed: 12/20/2022] Open
Abstract
Background Low conception rate (CR) despite insemination with morphologically normal spermatozoa is a common reproductive restraint that limits buffalo productivity. This accounts for a significant loss to the farmers and the dairy industry, especially in agriculture-based economies. The immune-related proteins on the sperm surface are known to regulate fertility by assisting the spermatozoa in their survival and performance in the female reproductive tract (FRT). Regardless of their importance, very few studies have specifically catalogued the buffalo sperm surface proteome. The study was designed to determine the identity of sperm surface proteins and to ascertain if the epididymal expressed beta-defensins (BDs), implicated in male fertility, are translated and applied onto buffalo sperm surface along with other immune-related proteins. Results The raw mass spectra data searched against an in-house generated proteome database from UniProt using Comet search engine identified more than 300 proteins on the ejaculated buffalo sperm surface which were bound either by non-covalent (ionic) interactions or by a glycosylphosphatidylinositol (GPI) anchor. The singular enrichment analysis (SEA) revealed that most of these proteins were extracellular with varied binding activities and were involved in either immune or reproductive processes. Flow cytometry using six FITC-labelled lectins confirmed the prediction of glycosylation of these proteins. Several beta-defensins (BDs), the anti-microbial peptides including the BuBD-129 and 126 were also identified amongst other buffalo sperm surface proteins. The presence of these proteins was subsequently confirmed by RT-qPCR, immunofluorescence and in vitro fertilization (IVF) experiments. Conclusions The surface of the buffalo spermatozoa is heavily glycosylated because of the epididymal secreted (glyco) proteins like BDs and the GPI-anchored proteins (GPI-APs). The glycosylation pattern of buffalo sperm-surface, however, could be perturbed in the presence of elevated salt concentration or incubation with PI-PLC. The identification of numerous BDs on the sperm surface strengthens our hypothesis that the buffalo BDs (BuBDs) assist the spermatozoa either in their survival or in performance in the FRT. Our results suggest that BuBD-129 is a sperm-surface BD that could have a role in buffalo sperm function. Further studies elucidating its exact physiological function are required to better understand its role in the regulation of male fertility. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07640-z.
Collapse
Affiliation(s)
- Vipul Batra
- Animal Genomics Lab., Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Vanya Bhushan
- Proteomics and Molecular Biology Lab, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Syed Azmal Ali
- Proteomics and Molecular Biology Lab, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Parul Sarwalia
- Animal Genomics Lab., Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Ankit Pal
- Animal Genomics Lab., Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Seema Karanwal
- Animal Genomics Lab., Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Subhash Solanki
- Animal Genomics Lab., Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Arumugam Kumaresan
- Theriogenology Lab, SRS of National Dairy Research Institute, Bengaluru, India
| | - Rakesh Kumar
- Animal Genomics Lab., Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Tirtha Kumar Datta
- Animal Genomics Lab., Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India.
| |
Collapse
|
20
|
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
|
21
|
Alves MBR, Arruda RPD, Batissaco L, Garcia-Oliveros LN, Gonzaga VHG, Nogueira VJM, Almeida FDS, Pinto SCC, Andrade GM, Perecin F, da Silveira JC, Celeghini ECC. Changes in miRNA levels of sperm and small extracellular vesicles of seminal plasma are associated with transient scrotal heat stress in bulls. Theriogenology 2020; 161:26-40. [PMID: 33278692 DOI: 10.1016/j.theriogenology.2020.11.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
Scrotal heat stress affects spermatogenesis and impairs male fertility by increasing sperm morphological abnormalities, oxidative stress and DNA fragmentation. While sperm morpho-functional changes triggered by scrotal heat stress are well described, sperm molecular alterations remain unknown. Recently, spermatozoa were described as accumulating miRNAs during the last steps of spermatogenesis and through epididymis transit, mainly by communication with small extracellular vesicles (sEVs). Herein, the aim was to investigate the impact of scrotal heat stress in miRNAs profile of sperm, as well as, seminal plasma sEVs. Six Nelore bulls (Bos indicus) were divided into two groups: Control (CON; n = 3) and Scrotal Heat Stress (SHS; n = 3; scrotal heat stressed during 96 h by scrotal bags). The day that the scrotal bags were removed from SHS group was considered as D0 (Day zero). Seminal plasma sEVs were isolated from semen samples collected seven days after heat stress (D+7) to evaluate sEVs diameter, concentration, and 380 miRNA levels. Sperm morpho-functional features and profile of 380 miRNAs were evaluated from semen collected 21 days after heat stress (D+21). As a control, sEVs and sperm were analyzed seven days before heat stress (D-7). Only semen parameters that were not significantly different (P > 0.05) among bulls on D-7 were addressed on D+7 and D+21. While no alterations in diameter and concentration were detected in sEVs on D+7 between CON and SHS groups, three sEVs-miRNAs (miR-23b-5p, -489 and -1248) were down-regulated in SHS bulls compared to CON on D+7; other three (miR-126-5p, -656 and -1307) displayed a tendency (0.05 < P < 0.10) to be altered. Sperm oxidative stress was higher, and the level of 21 sperm miRNAs was altered (18 down-, 3 up-regulated) in SHS bulls compared to CON on D+21. Functional analysis indicated that target genes involved in transcription activation, as well as cell proliferation and differentiation were related to the 18 down-regulated sperm miRNAs (miR-9-5p, -15a, -18a, -20b, -30a-5p, -30b-5p, -30d, -30e-5p -34b, -34c, -106b, -126-5p, -146a, -191, -192, -200b, -335 and -449a). Thus, the scrotal heat stress probably impacted testicular and epididymis functions by reducing the levels of a substantial proportion of sEVs and sperm miRNAs. Our findings suggest that miR-126-5p was possibly trafficked between sEVs and sperm and provide new insights on the mechanism by which sperm acquire miRNAs in the last stages of spermatogenesis and sperm maturation in cattle.
Collapse
Affiliation(s)
- Maíra Bianchi Rodrigues Alves
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Rubens Paes de Arruda
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Leonardo Batissaco
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Laura Nataly Garcia-Oliveros
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Vitor Hugo Guilger Gonzaga
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Vinícius José Moreira Nogueira
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Flávia Dos Santos Almeida
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Sâmara Cristine Costa Pinto
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Gabriella Mamede Andrade
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Felipe Perecin
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Juliano Coelho da Silveira
- Department of Veterinary Medicine, School of Animal Science and Food Engineering, University of São Paulo, Pirassununga, São Paulo, Brazil.
| | - Eneiva Carla Carvalho Celeghini
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil.
| |
Collapse
|
22
|
Faisal K, Akbarsha MA. Role of aposomes and epididymosomes in sperm quality control: A light and transmission electron microscopic study in an experimental rat model. Andrologia 2020; 53:e13862. [PMID: 33108830 DOI: 10.1111/and.13862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/07/2020] [Accepted: 09/12/2020] [Indexed: 12/01/2022] Open
Abstract
The epididymis responds to adverse conditions of misshapen spermatozoa resulting from pathological changes or toxic insults by secretion of a dense matrix that segregates the latter for complete disintegration and dissolution. The objective of this study was to find the source of this matrix and the role-player of disintegration and dissolution of misshapen spermatozoa. We chose Wistar strain male rat model to tackle this issue, and the rats were administered with aflatoxin B1 for 55 days so as to increase the incidence of misshapen spermatozoa. At the end of the treatment, different segments of epididymis were processed for microscopic observations. We found that parallel with abundant misshapen spermatozoa in the epididymis the principal cells of the initial segment secrete enormous membrane-bound apical blebs called aposomes, which contain epididymosomes. The aposomes were found to coalesce so as for the content to merge and form a dense matrix that entangles the misshapen spermatozoa and segregates them from viable spermatozoa. The epididymosomes associate with the misshapen spermatozoa, and the latter is processed to disintegration and total dissolution. Therefore, we assign the role of segregation of misshapen spermatozoa from viable ones to the dense matrix of aposomes and their disintegration and dissolution to the epididymosomes.
Collapse
Affiliation(s)
- Kunnathodi Faisal
- Department of Animal Science, Bharathidasan University, Tiruchirappalli, India
| | - Mohammad Abdulkader Akbarsha
- Department of Animal Science, Bharathidasan University, Tiruchirappalli, India.,Department of Biotechnology, National College (Autonomous), Tiruchirappalli, India
| |
Collapse
|
23
|
Candenas L, Chianese R. Exosome Composition and Seminal Plasma Proteome: A Promising Source of Biomarkers of Male Infertility. Int J Mol Sci 2020; 21:E7022. [PMID: 32987677 PMCID: PMC7583765 DOI: 10.3390/ijms21197022] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/09/2020] [Accepted: 09/19/2020] [Indexed: 02/06/2023] Open
Abstract
Infertility has become a global health issue, with approximately 50% of infertility cases generated by disorders in male reproduction. Spermatozoa are conveyed towards female genital tracts in a safe surrounding provided by the seminal plasma. Interestingly, this dynamically changing medium is a rich source of proteins, essential not only for sperm transport, but also for its protection and maturation. Most of the seminal proteins are acquired by spermatozoa in transit through exosomes (epididymosomes and prostasomes). The high number of seminal proteins, the increasing knowledge of their origins and biological functions and their differential expression in the case of azoospermia, asthenozoospermia, oligozoospermia and teratozoospermia or other conditions of male infertility have allowed the identification of a wide variety of biomarker candidates and their involvement in biological pathways, thus to strongly suggest that the proteomic landscape of seminal plasma may be a potential indicator of sperm dysfunction. This review summarizes the current knowledge in seminal plasma proteomics and its potentiality as a diagnostic tool in different degrees of male infertility.
Collapse
Affiliation(s)
- Luz Candenas
- Instituto de Investigaciones Químicas, CSIC, Avenida Américo Vespucio 49, 41092 Sevilla, Spain;
| | - Rosanna Chianese
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, via Costantinopoli 16, 80138 Napoli, Italy
| |
Collapse
|
24
|
Nätt D, Öst A. Male reproductive health and intergenerational metabolic responses from a small RNA perspective. J Intern Med 2020; 288:305-320. [PMID: 32415866 DOI: 10.1111/joim.13096] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/10/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023]
Abstract
The world has recently experienced a decline in male reproductive (e.g. sperm counts and motility) and metabolic (e.g. obesity and diabetes) health. Accumulated evidence from animal models also shows that the metabolic health of the father may influence the metabolic health in his offspring. Vectors for such paternal intergenerational metabolic responses (IGMRs) involve small noncoding RNAs (sncRNAs) that often increase in spermatozoa during the last days of maturation in the epididymis. We and others have shown that the metabolic state - depending on factors such as diet, obesity and physical exercise - may affect sperm quality and sperm sncRNA. Together, this suggests that there are overlapping aetiologies between the male metabolic syndrome, male factor infertility and intergenerational responses. In this review, we present a theoretical framework for an overlap of these aetiologies by exploring the advances in our understanding of the roles of sncRNA in spermatogenesis and offspring development. A special focus will lie on novel findings about tRNA-derived small RNA (tsRNA), rRNA-derived small RNA (rsRNA) and small mitochondrial RNA (mitoRNA), and their emerging roles in intergenerational metabolic and reproductive health.
Collapse
Affiliation(s)
- D Nätt
- From the, Department of Clinical and Experimental Medicine, Linköping University, Linkoping, Sweden
| | - A Öst
- From the, Department of Clinical and Experimental Medicine, Linköping University, Linkoping, Sweden
| |
Collapse
|
25
|
Leahy T, Rickard JP, Pini T, Gadella BM, Graaf SP. Quantitative Proteomic Analysis of Seminal Plasma, Sperm Membrane Proteins, and Seminal Extracellular Vesicles Suggests Vesicular Mechanisms Aid in the Removal and Addition of Proteins to the Ram Sperm Membrane. Proteomics 2020; 20:e1900289. [DOI: 10.1002/pmic.201900289] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 04/11/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Tamara Leahy
- School of Life and Environmental SciencesFaculty of ScienceUniversity of Sydney Sydney New South Wales 2006 Australia
| | - Jessica P. Rickard
- School of Life and Environmental SciencesFaculty of ScienceUniversity of Sydney Sydney New South Wales 2006 Australia
| | - Taylor Pini
- Colorado Center for Reproductive Medicine Lone Tree Colorado 80124 USA
| | - Bart M. Gadella
- Department of Farm Animal Health and Department of Biochemistry and Cell BiologyFaculty of Veterinary MedicineUtrecht University Yalelaan 2, CM Utrecht 3584 The Netherlands
| | - Simon P. Graaf
- School of Life and Environmental SciencesFaculty of ScienceUniversity of Sydney Sydney New South Wales 2006 Australia
| |
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
|
Touré A. Importance of SLC26 Transmembrane Anion Exchangers in Sperm Post-testicular Maturation and Fertilization Potential. Front Cell Dev Biol 2019; 7:230. [PMID: 31681763 PMCID: PMC6813192 DOI: 10.3389/fcell.2019.00230] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/26/2019] [Indexed: 12/17/2022] Open
Abstract
In mammals, sperm cells produced within the testis are structurally differentiated but remain immotile and are unable to fertilize the oocyte unless they undergo a series of maturation events during their transit in the male and female genital tracts. This post-testicular functional maturation is known to rely on the micro-environment of both male and female genital tracts, and is tightly controlled by the pH of their luminal milieus. In particular, within the epididymis, the establishment of a low bicarbonate (HCO3–) concentration contributes to luminal acidification, which is necessary for sperm maturation and subsequent storage in a quiescent state. Following ejaculation, sperm is exposed to the basic pH of the female genital tract and bicarbonate (HCO3–), calcium (Ca2+), and chloride (Cl–) influxes induce biochemical and electrophysiological changes to the sperm cells (cytoplasmic alkalinization, increased cAMP concentration, and protein phosphorylation cascades), which are indispensable for the acquisition of fertilization potential, a process called capacitation. Solute carrier 26 (SLC26) members are conserved membranous proteins that mediate the transport of various anions across the plasma membrane of epithelial cells and constitute important regulators of pH and HCO3– concentration. Most SLC26 members were shown to physically interact and cooperate with the cystic fibrosis transmembrane conductance regulator channel (CFTR) in various epithelia, mainly by stimulating its Cl– channel activity. Among SLC26 members, the function of SLC26A3, A6, and A8 were particularly investigated in the male genital tract and the sperm cells. In this review, we will focus on SLC26s contributions to ionic- and pH-dependent processes during sperm post-testicular maturation. We will specify the current knowledge regarding their functions, based on data from the literature generated by means of in vitro and in vivo studies in knock-out mouse models together with genetic studies of infertile patients. We will also discuss the limits of those studies, the current research gaps and identify some key points for potential developments in this field.
Collapse
Affiliation(s)
- Aminata Touré
- INSERM U1016, Centre National de la Recherche Scientifique, UMR 8104, Institut Cochin, Université de Paris, Paris, France
| |
Collapse
|
28
|
Murdica V, Cermisoni GC, Zarovni N, Salonia A, Viganò P, Vago R. Proteomic analysis reveals the negative modulator of sperm function glycodelin as over-represented in semen exosomes isolated from asthenozoospermic patients. Hum Reprod 2019; 34:1416-1427. [DOI: 10.1093/humrep/dez114] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/27/2019] [Indexed: 12/11/2022] Open
Abstract
ABSTRACT
STUDY QUESTION
Are there differences in the proteomic profile of exosomes isolated from seminal plasma of normozoospermic (NSP) and severe asthenozoospermic (SA) men, potentially contributing to sperm features?
SUMMARY ANSWER
A relevant group of proteins known to positively regulate sperm functions were over-represented in seminal exosomes of NSP men, i.e. cysteine-rich secretory protein-1 (CRISP1), while the inhibitory protein glycodelin was enriched in exosomes of SA subjects.
WHAT IS KNOWN ALREADY
Exosomes are secreted along the male reproductive tract and are thought to be involved in spermatozoa maturation and function. Ejaculated spermatozoa are still able to capture exosomes; exosomes of NSP individuals improve sperm motility and prompt capacitation, while exosomes of SA men fail to exert similar features.
STUDY DESIGN, SIZE, DURATION
Semen samples from NSP and SA men, aged 18 to 55 and registered at a single IVF center, were considered for this study project. Subjects were subdivided into three groups: a discovery cohort (five NSP men and six SA patients), a validation cohort (seven NSP and seven SA men) and the ‘glycodelin analysis’ cohort (20 NSP and 37 SA men). Exosomes were purified from semen of every participant.
PARTICIPANTS/MATERIALS, SETTING, METHODS
Exosomes were characterized by nanoparticle tracking analysis, transmission electron microscopy and western blot. Comprehensive proteomics analysis of the exosomal proteome was performed by nanoscale liquid chromatographic tandem mass spectrometry analysis. Funrich software was used to determine statistical enrichment of pathways, networks and Gene Ontology terms of the identified proteins. Validation of differentially expressed proteins was performed through ELISA and western blot analysis.
MAIN RESULTS AND THE ROLE OF CHANCE
The comprehensive proteomic analysis identified a total of 2138 proteins for both groups. There were 89 proteins found to be differentially expressed in exosomes of NSP versus SA subjects, of which 37 were increased in the NSP group and 52 were increased in the SA group. One-third of the exosomes-associated proteins highly expressed in NSP samples were involved in the reproductive process; conversely, the over-expressed proteins in exosomes of SA samples were not functionally specific. Quantitative data were confirmed on seminal exosomes from different cohorts of subjects.
LARGE SCALE DATA
N/A
LIMITATIONS, REASONS FOR CAUTION
Transfer of the proteins from exosomes to spermatozoa has been only partially demonstrated and up-take mechanisms are still poorly defined.
WIDER IMPLICATIONS OF THE FINDINGS
Seminal exosomes carry proteins that are potentially able to either favour or inhibit the reproductive process in humans. A better understanding of these phenomena might pave the way for novel intervention measures in terms of male infertility.
STUDY FUNDING/COMPETING INTEREST(S)
This study was funded by the Italian Ministry of Health through an Institution Seed Grant. None of the authors has any competing interests.
Collapse
Affiliation(s)
- Valentina Murdica
- Urological Research Institute, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Greta Chiara Cermisoni
- Centro Scienze Natalità, Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | | | - Andrea Salonia
- Urological Research Institute, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milano, Italy
- Università Vita-Salute San Raffaele, Milano, Italy
| | - Paola Viganò
- Reproductive Sciences Laboratory, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Riccardo Vago
- Urological Research Institute, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milano, Italy
- Università Vita-Salute San Raffaele, Milano, Italy
| |
Collapse
|
29
|
Efficient isolation, biophysical characterisation and molecular composition of extracellular vesicles secreted by primary and immortalised cells of reproductive origin. Theriogenology 2019; 135:121-137. [PMID: 31207473 DOI: 10.1016/j.theriogenology.2019.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/25/2019] [Accepted: 06/04/2019] [Indexed: 12/27/2022]
Abstract
Effective communication between the maternal reproductive tract, gametes and the pre-implantation embryo is essential for the successful establishment of pregnancy. Recent studies have recognised extracellular vesicles (EVs) as potent vehicles for intercellular communication, potentially via their transport of microRNAs (miRNAs). The aim of the current investigation was to determine the size, concentration and electrical surface properties (zeta potential) of EVs secreted by; (1) primary cultures of porcine oviductal epithelial cells (POECs) from the isthmus and ampullary regions of the female reproductive tract; (2) Ishikawa and RL95-2 human endometrial epithelial cell line cultures; and (3) the non-reproductive epithelial cell line HEK293T. In addition, this study investigated whether EVs secreted by POECs contained miRNAs. All cell types were cultured in EV-depleted medium for 24 or 48 h. EVs were successfully isolated from conditioned culture media using size exclusion chromatography. Nanoparticle tracking analysis (NTA) was performed to evaluate EV size, concentration and zeta potential. QRT-PCR was performed to quantify the expression of candidate miRNAs (miR-103, let-7a, miR-19a, miR-203, miR-126, miR-19b, RNU44, miR-92, miR-196a, miR-326 and miR-23a). NTA confirmed the presence of EVs with diameters of 50-150 nm in all cell types. EV size distribution was significantly different between cell types after 24 and 48 h of cell culture and the concentration of EVs secreted by POECs and Ishikawa cells was also time dependent. The distribution of EVs with specific electrokinetic potential measurements varied between cell types, indicating that EVs of differing cellular origin have varied membrane components. In addition, EVs secreted by POECs exhibited significantly different time dependant changes in zeta potential. QRT-PCR confirmed the presence of miR-103, let-7a, miR-19a, miR-203, miR-126, and miR-19b in EVs secreted by POECs (CT ≥ 29). Bioinformatics analysis suggests that these miRNAs are involved in cell proliferation, innate immune responses, apoptosis and cellular migration. In conclusion, reproductive epithelial cells secrete distinct populations of EVs containing miRNAs, which potentially act in intercellular communication in order to modulate the periconception events leading to successful establishment of pregnancy.
Collapse
|
30
|
Trigg NA, Eamens AL, Nixon B. The contribution of epididymosomes to the sperm small RNA profile. Reproduction 2019; 157:R209-R223. [DOI: 10.1530/rep-18-0480] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/18/2019] [Indexed: 12/16/2022]
Abstract
It is now well established that mature spermatozoa harbour a rich and diverse profile of small non-protein-coding regulatory RNAs (sRNAs). There is also growing appreciation that this sRNA profile displays considerable plasticity, being altered in response to paternal exposure to a variety of environmental stressors. Coupled with evidence that upon delivery to the oocyte at the moment of fertilisation, sperm-borne sRNAs are able to influence both early embryonic development and the subsequent health of the offspring, there is now interest in both the timing and degree of change in the composition of the sRNA cargo of sperm. Models in which such epigenetic changes are linked to the spermatogenic cycle are seemingly incompatible with the lack of overt phenotypic changes in the spermatozoa of affected males. Rather, there is mounting consensus that such changes are imposed on sperm during their transit and storage within the epididymis, a protracted developmental window that takes place over several weeks. Notably, since spermatozoa are rendered transcriptionally and translationally silent during their development in the testes, it is most likely that the epididymis-documented alterations to the sperm sRNA profile are driven extrinsically, with a leading candidate being epididymosomes: small membrane enclosed extracellular vesicles that encapsulate a complex macromolecular cargo of proteins and RNAs, including the sRNAs. Here, we review the role of epididymosome–sperm communication in contributing to the establishment of the sperm sRNA profile during their epididymal transit.
Collapse
|
31
|
O'Flaherty C. Orchestrating the antioxidant defenses in the epididymis. Andrology 2019; 7:662-668. [PMID: 31044545 DOI: 10.1111/andr.12630] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/26/2018] [Accepted: 03/29/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND During the post-testicular maturation that occurs in the epididymis, spermatozoa need to face biochemical and morphological changes that may make them vulnerable to oxidative damage. During spermatogenesis and the epididymal maturation, the spermatozoon acquires antioxidant enzymes needed to face possible increases of reactive oxygen species (ROS) produced by its own aerobic metabolism but also due to ROS produced in high quantities by abnormal spermatozoa. OBJECTIVES Provide an up-to-date review of the enzymatic antioxidant system in the epididymis. MATERIAL AND METHODS A thorough literature review was performed for papers concerning the players of the antioxidant defenses in the epididymis. RESULTS The antioxidant system in the epididymis is composed by superoxide dismutases, catalase, glutathione peroxidases, peroxiredoxins, glutathione-S-transferases, thioredoxins and thioredoxin reductase. They work together to maintain low levels of ROS during the epididymal maturation. Knockout models revealed that the absence of one of the enzyme impact sperm quality affecting a variety of proteins involved in motility, the ability to fertilize oocyte, and promotes oxidative damage to the sperm DNA. DISCUSSION AND CONCLUSIONS These findings suggest that each enzyme is playing a specific role, and in most of the cases, no compensatory mechanisms are put in place when one enzyme is absent. This review highlights the different antioxidant enzymes in the epididymis and their role during maturation of the spermatozoon.
Collapse
Affiliation(s)
- C O'Flaherty
- Departments of Surgery (Urology Division) and Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada.,The Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
32
|
Zhou W, Stanger SJ, Anderson AL, Bernstein IR, De Iuliis GN, McCluskey A, McLaughlin EA, Dun MD, Nixon B. Mechanisms of tethering and cargo transfer during epididymosome-sperm interactions. BMC Biol 2019; 17:35. [PMID: 30999907 PMCID: PMC6474069 DOI: 10.1186/s12915-019-0653-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 04/04/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The mammalian epididymis is responsible for the provision of a highly specialized environment in which spermatozoa acquire functional maturity and are subsequently stored in preparation for ejaculation. Making important contributions to both processes are epididymosomes, small extracellular vesicles released from the epididymal soma via an apocrine secretory pathway. While considerable effort has been focused on defining the cargo transferred between epididymosomes and spermatozoa, comparatively less is known about the mechanistic basis of these interactions. To investigate this phenomenon, we have utilized an in vitro co-culture system to track the transfer of biotinylated protein cargo between mouse epididymosomes and recipient spermatozoa isolated from the caput epididymis; an epididymal segment that is of critical importance for promoting sperm maturation. RESULTS Our data indicate that epididymosome-sperm interactions are initiated via tethering of the epididymosome to receptors restricted to the post-acrosomal domain of the sperm head. Thereafter, epididymosomes mediate the transfer of protein cargo to spermatozoa via a process that is dependent on dynamin, a family of mechanoenzymes that direct intercellular vesicle trafficking. Notably, upon co-culture of sperm with epididymosomes, dynamin 1 undergoes a pronounced relocation between the peri- and post-acrosomal domains of the sperm head. This repositioning of dynamin 1 is potentially mediated via its association with membrane rafts and ideally locates the enzyme to facilitate the uptake of epididymosome-borne proteins. Accordingly, disruption of membrane raft integrity or pharmacological inhibition of dynamin both potently suppress the transfer of biotinylated epididymosome proteins to spermatozoa. CONCLUSION Together, these data provide new mechanistic insight into epididymosome-sperm interactions with potential implications extending to the manipulation of sperm maturation for the purpose of fertility regulation.
Collapse
Affiliation(s)
- Wei Zhou
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Simone J Stanger
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Amanda L Anderson
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Ilana R Bernstein
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Geoffry N De Iuliis
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Adam McCluskey
- Priority Research Centre for Chemical Biology, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Eileen A McLaughlin
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,School of Biological Sciences, University of Auckland, Auckland, 1142, New Zealand.,Faculty of Science and Technology, University of Canberra, Bruce, ACT, 2617, Australia
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Cancer Research Program, New Lambton Heights, NSW, 2305, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
33
|
Nixon B, De Iuliis GN, Hart HM, Zhou W, Mathe A, Bernstein IR, Anderson AL, Stanger SJ, Skerrett-Byrne DA, Jamaluddin MFB, Almazi JG, Bromfield EG, Larsen MR, Dun MD. Proteomic Profiling of Mouse Epididymosomes Reveals their Contributions to Post-testicular Sperm Maturation. Mol Cell Proteomics 2019; 18:S91-S108. [PMID: 30213844 PMCID: PMC6427233 DOI: 10.1074/mcp.ra118.000946] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/28/2018] [Indexed: 01/31/2023] Open
Abstract
The functional maturation of spermatozoa that is necessary to achieve fertilization occurs as these cells transit through the epididymis, a highly specialized region of the male reproductive tract. A defining feature of this maturation process is that it occurs in the complete absence of nuclear gene transcription or de novo, protein translation in the spermatozoa. Rather, it is driven by sequential interactions between spermatozoa and the complex external milieu in which they are bathed within lumen of the epididymal tubule. A feature of this dynamic microenvironment are epididymosomes, small membrane encapsulated vesicles that are secreted from the epididymal soma. Herein, we report comparative proteomic profiling of epididymosomes isolated from different segments of the mouse epididymis using multiplexed tandem mass tag (TMT) based quantification coupled with high resolution LC-MS/MS. A total of 1640 epididymosome proteins were identified and quantified via this proteomic method. Notably, this analysis revealed pronounced segment-to-segment variation in the encapsulated epididymosome proteome. Thus, 146 proteins were identified as being differentially accumulated between caput and corpus epididymosomes, and a further 344 were differentially accumulated between corpus and cauda epididymosomes (i.e., fold change of ≤ -1.5 or ≥ 1.5; p, < 0.05). Application of gene ontology annotation revealed a substantial portion of the epididymosome proteins mapped to the cellular component of extracellular exosome and to the biological processes of transport, oxidation-reduction, and metabolism. Additional annotation of the subset of epididymosome proteins that have not previously been identified in exosomes revealed enrichment of categories associated with the acquisition of sperm function (e.g., fertilization and binding to the zona pellucida). In tandem with our demonstration that epididymosomes are able to convey protein cargo to the head of maturing spermatozoa, these data emphasize the fundamental importance of epididymosomes as key elements of the epididymal microenvironment responsible for coordinating post-testicular sperm maturation.
Collapse
Affiliation(s)
- Brett Nixon
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Geoffry N De Iuliis
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Hanah M Hart
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Wei Zhou
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Andrea Mathe
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia;; School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Ilana R Bernstein
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Amanda L Anderson
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Simone J Stanger
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - David A Skerrett-Byrne
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - M Fairuz B Jamaluddin
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia;; Hunter Medical Research Institute, Cancer Research Program, New Lambton Heights, NSW 2305, Australia
| | - Juhura G Almazi
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia;; Hunter Medical Research Institute, Cancer Research Program, New Lambton Heights, NSW 2305, Australia
| | - Elizabeth G Bromfield
- From the ‡Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia;; Hunter Medical Research Institute, Cancer Research Program, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
34
|
O'Flaherty C, Matsushita-Fournier D. Reactive oxygen species and protein modifications in spermatozoa. Biol Reprod 2018; 97:577-585. [PMID: 29025014 DOI: 10.1093/biolre/iox104] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 09/11/2017] [Indexed: 02/07/2023] Open
Abstract
Cellular response to reactive oxygen species (ROS) includes both reversible redox signaling and irreversible nonenzymatic reactions which depend on the nature and concentration of the ROS involved. Changes in thiol/disulfide pairs affect protein conformation, enzymatic activity, ligand binding, and protein-protein interactions. During spermatogenesis and epididymal maturation, there are ROS-dependent modifications of the sperm chromatin and flagellar proteins.The spermatozoon is regulated by redox mechanisms to acquire fertilizing ability. For this purpose, controlled amounts of ROS are necessary to assure sperm activation (motility and capacitation). Modifications of the thiol groups redox status of sperm proteins are needed for spermatozoon to achieve fertilizing ability. However, when ROS are produced at high concentrations, the established oxidative stress promotes pathological changes affecting sperm function and leading to infertility. Sperm proteins are sensitive to high levels of ROS and suffer modifications that impact on motility, capacitation, and the ability of the spermatozoon to recognize and bind to the zona pellucida and damage of sperm DNA. Thiol oxidation, tyrosine nitration, and S-glutathionylation are highlighted in this review as significant redox-dependent protein modifications associated with impairment of sperm function and alteration of paternal genome leading to infertility. Peroxiredoxins, the primary antioxidant protection in spermatozoa, are affected by most of the protein modifications described in this review. They play a significant role in both physiological and pathological processes in mammalian spermatozoa.
Collapse
Affiliation(s)
- Cristian O'Flaherty
- Department of Surgery (Urology Division), McGill University, Montréal, Québec, Canada.,Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montréal, Québec, Canada.,The Research Institute, McGill University Health Centre, Montréal, Québec, Canada
| | - David Matsushita-Fournier
- Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montréal, Québec, Canada.,The Research Institute, McGill University Health Centre, Montréal, Québec, Canada
| |
Collapse
|
35
|
Liu Y, O'Flaherty C. In vivo oxidative stress alters thiol redox status of peroxiredoxin 1 and 6 and impairs rat sperm quality. Asian J Androl 2017; 19:73-79. [PMID: 26823067 PMCID: PMC5227679 DOI: 10.4103/1008-682x.170863] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Oxidative stress, the imbalance between the production of reactive oxygen species (ROS) and antioxidant activity is a major culprit of male infertility. Peroxiredoxins (PRDXs) are major antioxidant enzymes of mammalian spermatozoa and are thiol oxidized and inactivated by ROS in a dose-dependent manner. Their deficiency and/or inactivation have been associated with men infertility. The aim of this study was to elucidate the impact of oxidative stress, generated by the in vivo tert-butyl hydroperoxide (tert-BHP) treatment on rat epididymal spermatozoa during their maturation process. Adult Sprague-Dawley males were treated with μmoles tert-BHP/kg or saline (control) per day intraperitoneal for 15 days. Lipid peroxidation (2-thibarbituric acid reactive substances assay), total amount and thiol oxidation of PRDXs along with the total amount of superoxide dismutase (SOD), motility and DNA oxidation (8-hydroxy-deoxyguanosine) were determined in epididymal spermatozoa. Total amount of PRDXs and catalase and thiol oxidation of PRDXs were determined in caput and cauda epididymis. While animals were not affected by treatment, their epididymal spermatozoa have decreased motility, increased levels of DNA oxidation and lipid peroxidation along with increased PRDXs (and not SOD) amounts. Moreover, sperm PRDXs were highly thiol oxidized. There was a differential regulation in the expression of PRDX1 and PRDX6 in the epididymis that suggests a segment-specific role for PRDXs. In conclusion, PRDXs are increased in epididymal spermatozoa in an attempt to fight against the oxidative stress generated by tert-BHP in the epididymis. These findings highlight the role of PRDXs in the protection of sperm function and DNA integrity during epididymal maturation.
Collapse
Affiliation(s)
- Yannan Liu
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.,Department of Surgery (Urology Division), McGill University, Montreal, QC H4A 3J1, Canada
| | - Cristian O'Flaherty
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.,Department of Surgery (Urology Division), McGill University, Montreal, QC H4A 3J1, Canada.,Department of Obstetrics and Gynecology, McGill University, Montreal, QC H4A 3J1, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
36
|
Madison MN, Welch JL, Okeoma CM. Isolation of Exosomes from Semen for in vitro Uptake and HIV-1 Infection Assays. Bio Protoc 2017; 7:e2216. [PMID: 28660234 DOI: 10.21769/bioprotoc.2216] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Exosomes are membranous extracellular nanovesicles of endocytic origin. Exosomes are known to carry host and pathogen-derived genomic, proteomic, lipidomic cargos and other extraneous molecules. Exosomes are secreted by diverse cell types into the extracellular milieu and are subsequently internalized by recipient neighboring or distal cells. Upon internalization, exosomes condition recipient cells by donating their cargos and/or activating various signal transduction pathways, consequently regulating physiological and pathophysiological processes. Exosomes facilitate intercellular communication, modulate cellular phenotype, and regulate microbial pathogenesis. We have previously shown that semen exosomes (SE) inhibit HIV-1 replication in various cell types. Here, we describe detailed protocols for characterizing SE. This protocol can be adapted or modified and used for evaluation of other extracellular vesicles of interest.
Collapse
Affiliation(s)
- Marisa N Madison
- Department of Mathematics and Natural Sciences, Miami Dade College, Homestead, USA
| | - Jennifer L Welch
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Chioma M Okeoma
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, USA.,Interdisciplinary Program in Molecular and Cellular Biology, University of Iowa, Iowa City, USA
| |
Collapse
|
37
|
Gelatin Binding Proteins in Reproductive Physiology. Indian J Microbiol 2016; 56:383-393. [PMID: 27784933 DOI: 10.1007/s12088-016-0618-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 08/25/2016] [Indexed: 10/21/2022] Open
Abstract
In order to advance the assisted reproductive technologies used in animals and human beings, it is important to accumulate basic informations about underlying molecular mechanisms that shape the biological processes of reproduction. From within seminal plasma, proteins perform a wide variety of distinct functions that regulate major reproductive events such as fertilization. The ability of such proteins to bind and interact with different antagonistic ions and biomolecules such as polysaccharides, lipids, and other proteins present in the male and female reproductive tract define these capabilities. Over the last two decades, extensive work has been undertaken in an attempt to define the role of seminal plasma proteins, of which, Gelatin binding proteins (GBPs) represent a large family. GBPs comprise of known group of Bovine seminal plasma (BSP) protein family, matrix metallo proteinases (MMP 2 and MMP 9) and fibronectin, which have been widely studied. The presence of a type II repeat is a characteristic feature of GBPs, which is similar in structure to the fibronectin type II domain (fn2), which has ability to bind multiple ligands including gelatin, glycosaminoglycans, choline phospholipids, and lipoproteins. Two fn2 domains are present within the BSP protein family, while, three fn2 domains are found in gelatinases (MMP-2 and MMP9), and ELSPBP1 (Epididymosomes Transfer Epididymal Sperm Binding Protein 1) contains four long fn2 domains. For the most part BSP proteins are exclusively expressed in seminal vesicles although mBSPH1, mBSPH2 and hBSPH1 are all expressed in the epididymis. The expression of gelatinases has been demonstrated in several organs and tissues such as the prostate, testis, epididymis, ovary, human placenta, cervix and endometrial wall. This review intends to bring current updates on the role of GBPs in reproductive physiology to light, which may act as basis for future studies on GBPs.
Collapse
|
38
|
Characterisation of mouse epididymosomes reveals a complex profile of microRNAs and a potential mechanism for modification of the sperm epigenome. Sci Rep 2016; 6:31794. [PMID: 27549865 PMCID: PMC4994100 DOI: 10.1038/srep31794] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/26/2016] [Indexed: 12/18/2022] Open
Abstract
Recent evidence has shown that the sperm epigenome is vulnerable to dynamic modifications arising from a variety of paternal environment exposures and that this legacy can serve as an important determinant of intergenerational inheritance. It has been postulated that such exchange is communicated to maturing spermatozoa via the transfer of small non-protein-coding RNAs (sRNAs) in a mechanism mediated by epididymosomes; small membrane bound vesicles released by the soma of the male reproductive tract (epididymis). Here we confirm that mouse epididymosomes encapsulate an impressive cargo of >350 microRNAs (miRNAs), a developmentally important sRNA class, the majority (~60%) of which are also represented by the miRNA signature of spermatozoa. This includes >50 miRNAs that were found exclusively in epididymal sperm and epididymosomes, but not in the surrounding soma. We also documented substantial changes in the epididymosome miRNA cargo, including significant fold changes in almost half of the miRNAs along the length of the epididymis. Finally, we provide the first direct evidence for the transfer of several prominent miRNA species between mouse epididymosomes and spermatozoa to afford novel insight into a mechanism of intercellular communication by which the sRNA payload of sperm can be selectively modified during their post-testicular maturation.
Collapse
|
39
|
Bernabò N, Agostino RD, Ordinelli A, Mattioli M, Barboni B. The maturation of murine spermatozoa membranes within the epididymis, a computational biology perspective. Syst Biol Reprod Med 2016; 62:299-308. [PMID: 27586844 DOI: 10.1080/19396368.2016.1205679] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
To become fertile, mammalian spermatozoa require completing a complex biochemical maturation that begins in the testis and ends within the female oviduct. Here, we paid attention to the events occurring at the membrane level during the epididymal transit. Indeed, in the epididymis, the molecular composition and the physical-chemical proprieties of sperm membranes markedly change, with functional cross talking among the spermatozoa, the epithelium, and the luminal content (particularly the epididymosomes). To study this process, we undertook a biological networks study, representing the involved molecules as nodes and their interactions as links. The analysis of network topology revealed that it has a scale free and small world architecture and it is robust against random failure. That assures a fast and efficient transmission of information and it leads to identifying the molecules exerting a higher level of control on the system, among which cholesterol plays a pivotal role. The reactome enrichment analysis allowed the reconstruction of the biochemical pathways involved in sperm epididymal maturation and STRING analysis permitted the identification of molecular events possibly involved in that process. In conclusion, this approach allows inferring interesting information, thus contributing to the knowledge on this process and suggesting staring points for further research.
Collapse
Affiliation(s)
- Nicola Bernabò
- a Faculty of Veterinary Medicine , University of Teramo , Teramo , Italy
| | | | | | - Mauro Mattioli
- a Faculty of Veterinary Medicine , University of Teramo , Teramo , Italy
| | - Barbara Barboni
- a Faculty of Veterinary Medicine , University of Teramo , Teramo , Italy
| |
Collapse
|
40
|
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
|
41
|
Machtinger R, Laurent LC, Baccarelli AA. Extracellular vesicles: roles in gamete maturation, fertilization and embryo implantation. Hum Reprod Update 2015; 22:182-93. [PMID: 26663221 DOI: 10.1093/humupd/dmv055] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 11/09/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are membrane-bound vesicles, found in biofluids, that carry and transfer regulatory molecules, such as microRNAs (miRNAs) and proteins, and may mediate intercellular communication between cells and tissues. EVs have been isolated from a wide variety of biofluids, including plasma, urine, and, relevant to this review, seminal, follicular and uterine luminal fluid. We conducted a systematic search of the literature to review and present the currently available evidence on the possible roles of EVs in follicular growth, resumption of oocyte development and maturation (meiosis), sperm maturation, fertilization and embryo implantation. METHODS MEDLINE, Embase and Web of Science databases were searched using keywords pertaining to EVs, including 'extracellular vesicles', 'microvesicles', 'microparticles' and 'exosomes', combined with a range of terms associated with the period of development between fertilization and implantation, including 'oocyte', 'sperm', 'semen', 'fertilization', 'implantation', 'embryo', 'follicular fluid', 'epididymal fluid' and 'seminal fluid'. Relevant research articles published in English (both animal and human studies) were reviewed with no restrictions on publication date (i.e. from earliest database dates to July 2015). References from these articles were used to obtain additional articles. RESULTS A total of 1556 records were retrieved from the three databases. After removing duplicates and irrelevant titles, we reviewed the abstracts of 201 articles, which included 92 relevant articles. Both animal and human studies unequivocally identified various types of EVs in seminal, follicular and ULFs. Several studies provided evidence for the roles of EVs in these biofluids. In men, EVs in seminal fluid were linked with post-testicular sperm maturation, including sperm motility acquisition and reduction of oxidative stress. In women, EVs in follicular fluid were shown to contain miRNAs with potential roles in follicular growth, resumption of oocyte meiosis, steroidogenesis and prevention of polyspermy after fertilization. EVs were also detected in the media of cultured embryos, suggesting that EVs released from embryos and the uterus may mediate embryo-endometrium cross-talk during implantation. It is important to note that many of the biologically plausible functions of EVs in reproduction discussed in the current literature have not yet been substantiated by conclusive experimental evidence. CONCLUSIONS A detailed understanding of the contributions of EVs in the series of events from gametogenesis to fertilization and then on to implantation, in both normal and pathological cases, may enable the development of valuable tools to advance reproductive health. Because of the early stage of the field, it is unsurprising that the current literature includes not only growing experimental evidence, but also as-yet unproven hypotheses pertaining to the roles of EVs in key reproductive processes. In this review, we present a comprehensive survey of the rapidly expanding literature on this subject, highlighting both relevant findings and gaps in knowledge.
Collapse
Affiliation(s)
- Ronit Machtinger
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Sheba Medical Center and Tel-Aviv University, Tel Hashomer 52561, Israel
| | - Louise C Laurent
- Department of Reproductive Medicine, Division of Maternal Fetal Medicine, University of California, San Diego, CA, USA
| | - Andrea A Baccarelli
- Departments of Environmental Health and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
42
|
Skerget S, Rosenow MA, Petritis K, Karr TL. Sperm Proteome Maturation in the Mouse Epididymis. PLoS One 2015; 10:e0140650. [PMID: 26556802 PMCID: PMC4640836 DOI: 10.1371/journal.pone.0140650] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 09/29/2015] [Indexed: 12/19/2022] Open
Abstract
In mammals, transit through the epididymis, which involves the acquisition, loss and modification of proteins, is required to confer motility and fertilization competency to sperm. The overall dynamics of maturation is poorly understood, and a systems level understanding of the complex maturation process will provide valuable new information about changes occurring during epididymal transport. We report the proteomes of sperm collected from the caput, corpus and cauda segments of the mouse epididymis, identifying 1536, 1720 and 1234 proteins respectively. This study identified 765 proteins that are present in sperm obtained from all three segments. We identified 1766 proteins that are potentially added (732) or removed (1034) from sperm during epididymal transit. Phenotypic analyses of the caput, corpus and cauda sperm proteomes identified 60 proteins that have known sperm phenotypes when mutated, or absent from sperm. Our analysis indicates that as much as one-third of proteins with known sperm phenotypes are added to sperm during epididymal transit. GO analyses revealed that cauda sperm are enriched for specific functions including sperm-egg recognition and motility, consistent with the observation that sperm acquire motility and fertilization competency during transit through the epididymis. In addition, GO analyses revealed that the immunity protein profile of sperm changes during sperm maturation. Finally, we identified components of the 26S proteasome, the immunoproteasome, and a proteasome activator in mature sperm.
Collapse
Affiliation(s)
- Sheri Skerget
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | - Matthew A Rosenow
- Center for Proteomics, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Konstantinos Petritis
- Center for Proteomics, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Timothy L Karr
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| |
Collapse
|
43
|
Abstract
Exosomes are membranous nanovesicles of endocytic origin that carry host and pathogen derived genomic, proteomic, and lipid cargos. Exosomes are secreted by most cell types into the extracellular milieu and are subsequently internalized by recipient cells. Upon internalization, exosomes condition recipient cells by donating their cargos and/or activating various signal transduction pathways, consequently regulating physiological and pathophysiological processes. The role of exosomes in viral pathogenesis, especially human immunodeficiency virus type 1 [HIV-1] is beginning to unravel. Recent research reports suggest that exosomes from various sources play important but different roles in the pathogenesis of HIV-1. From these reports, it appears that the source of exosomes is the defining factor for the exosomal effect on HIV-1. In this review, we will describe how HIV-1 infection is modulated by exosomes and in turn how exosomes are targeted by HIV-1 factors. Finally, we will discuss potentially emerging therapeutic options based on exosomal cargos that may have promise in preventing HIV-1 transmission.
Collapse
|
44
|
Oviduct binding ability of porcine spermatozoa develops in the epididymis and can be advanced by incubation with caudal fluid. Theriogenology 2015; 83:1502-13. [DOI: 10.1016/j.theriogenology.2015.01.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 01/27/2015] [Accepted: 01/28/2015] [Indexed: 11/23/2022]
|
45
|
Protein-Carbohydrate Interaction between Sperm and the Egg-Coating Envelope and Its Regulation by Dicalcin, a Xenopus laevis Zona Pellucida Protein-Associated Protein. Molecules 2015; 20:9468-86. [PMID: 26007194 PMCID: PMC6272592 DOI: 10.3390/molecules20059468] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/13/2015] [Indexed: 12/17/2022] Open
Abstract
Protein-carbohydrate interaction regulates multiple important processes during fertilization, an essential biological event where individual gametes undergo intercellular recognition to fuse and generate a zygote. In the mammalian female reproductive tract, sperm temporarily adhere to the oviductal epithelium via the complementary interaction between carbohydrate-binding proteins on the sperm membrane and carbohydrates on the oviductal cells. After detachment from the oviductal epithelium at the appropriate time point following ovulation, sperm migrate and occasionally bind to the extracellular matrix, called the zona pellucida (ZP), which surrounds the egg, thereafter undergoing the exocytotic acrosomal reaction to penetrate the envelope and to reach the egg plasma membrane. This sperm-ZP interaction also involves the direct interaction between sperm carbohydrate-binding proteins and carbohydrates within the ZP, most of which have been conserved across divergent species from mammals to amphibians and echinoderms. This review focuses on the carbohydrate-mediated interaction of sperm with the female reproductive tract, mainly the interaction between sperm and the ZP, and introduces the fertilization-suppressive action of dicalcin, a Xenopus laevis ZP protein-associated protein. The action of dicalcin correlates significantly with a dicalcin-dependent change in the lectin-staining pattern within the ZP, suggesting a unique role of dicalcin as an inherent protein that is capable of regulating the affinity between the lectin and oligosaccharides attached on its target glycoprotein.
Collapse
|
46
|
Akintayo A, Légaré C, Sullivan R. Dicarbonyl L-xylulose reductase (DCXR), a "moonlighting protein" in the bovine epididymis. PLoS One 2015; 10:e0120869. [PMID: 25815750 PMCID: PMC4376396 DOI: 10.1371/journal.pone.0120869] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/28/2015] [Indexed: 01/10/2023] Open
Abstract
During maturation and the acquisition of their fertilization potential, male germ cells are subjected to various sequential modifications that occur in the epididymis. Protein addition, reorganization or withdrawal, comprise some of these modifications. Dicarbonyl L-xylulose reductase (DCXR), a multifunctional protein involved in various enzymatic and protein interaction processes in different physiological systems, is one of the proteins added to spermatozoa in the epididymis. DCXR is a well-conserved protein with multiple characteristics including enzymatic activities and mediation of cell-cell interaction. In this study, we characterized the DCXR gene and protein expression in the bovine epididymis. Dicarbonyl L-xylulose reductase mRNA is differentially expressed in the caput, corpus, and cauda epididymide epithelial cells with a higher level observed in the cauda region. Tissue protein expression follows the same pattern as the corresponding mRNA expression with a cytoplasmic and apical distribution in the corpus and cauda epithelial cells, respectively. The protein can also be found with a nuclear localization in cauda epididymidis epithelial cells. Dicarbonyl L-xylulose reductase is secreted in the epididymis luminal compartment in the soluble fraction and is associated with microvesicular elements named epididymosomes. In spermatozoa, the DCXR protein was found in the cytoplasmic and membranous fractions. Expression of the DCXR protein is higher on caput spermatozoa but finally shows a weak detection in semen. These data describe DCXR in the bovine epididymis and reveal that its behavior differs from that found in humans. It seems that, in this model, the DCXR protein might have a questionable involvement in the fertilization process.
Collapse
Affiliation(s)
- Ayodélé Akintayo
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec, Département d’Obstétrique, Gynécologie et Reproduction, Université Laval, Faculté de Medicine, Québec, Canada
| | - Christine Légaré
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec, Département d’Obstétrique, Gynécologie et Reproduction, Université Laval, Faculté de Medicine, Québec, Canada
| | - Robert Sullivan
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec, Département d’Obstétrique, Gynécologie et Reproduction, Université Laval, Faculté de Medicine, Québec, Canada
- * E-mail:
| |
Collapse
|
47
|
Păunescu TG, Shum WWC, Huynh C, Lechner L, Goetze B, Brown D, Breton S. High-resolution helium ion microscopy of epididymal epithelial cells and their interaction with spermatozoa. Mol Hum Reprod 2014; 20:929-37. [PMID: 25015675 PMCID: PMC4172170 DOI: 10.1093/molehr/gau052] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/20/2014] [Accepted: 07/01/2014] [Indexed: 01/19/2023] Open
Abstract
We examined the rat and mouse epididymis using helium ion microscopy (HIM), a novel imaging technology that uses a scanning beam of He(+) ions to produce nanometer resolution images of uncoated biological samples. Various tissue fixation, sectioning and dehydration methods were evaluated for their ability to preserve tissue architecture. The cauda epididymidis was luminally perfused in vivo to remove most spermatozoa and the apical surface of the epithelial lining was exposed. Fixed epididymis samples were then subjected to critical point drying (CPD) and HIM. Apical stereocilia in principal cells and smaller apical membrane extensions in clear cells were clearly distinguishable in both rat and mouse epididymis using this technology. After perfusion with an activating solution containing CPT-cAMP, a permeant analog of cAMP, clear cells exhibited an increase in the number and size of membrane ruffles or microplicae. In contrast, principal cells did not exhibit detectable structural modifications. High-resolution HIM imaging clearly showed the ultrastructure of residual sperm cells, including the presence of concentric rings on the midpiece, and of cytoplasmic droplets in some spermatozoa. Close epithelium-sperm interactions were also detected. We found a number of sperm cells whose heads were anchored within the epididymal epithelium. In certain cases, the surface of the sperm cytoplasmic droplet was covered with vesicle-like structures whose size is consistent with that of epididymosomes. In conclusion, we describe here the first application of HIM technology to the study of the structure and morphology of the rodent epididymis. HIM technology represents a major imaging breakthrough that can be successfully applied to study the epididymis and spermatozoa, with the goal of advancing our understanding of their structure and function.
Collapse
Affiliation(s)
- Teodor G Păunescu
- Department of Medicine, Program in Membrane Biology and Division of Nephrology, Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Center, CPZN 8204, 185 Cambridge St, Boston, MA 02114, USA
| | - Winnie W C Shum
- Department of Medicine, Program in Membrane Biology and Division of Nephrology, Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Center, CPZN 8204, 185 Cambridge St, Boston, MA 02114, USA Present address: School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | | | | | | | - Dennis Brown
- Department of Medicine, Program in Membrane Biology and Division of Nephrology, Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Center, CPZN 8204, 185 Cambridge St, Boston, MA 02114, USA
| | - Sylvie Breton
- Department of Medicine, Program in Membrane Biology and Division of Nephrology, Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Center, CPZN 8204, 185 Cambridge St, Boston, MA 02114, USA
| |
Collapse
|
48
|
He J, Li X, Luo D, Zhang C, Hu S, Li X. A new animal bioreactor for producing pharmaceutical proteins. Acta Biochim Biophys Sin (Shanghai) 2014; 46:826-828. [PMID: 25033830 DOI: 10.1093/abbs/gmu062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jinshui He
- Department of Pediatrics, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, China
| | - Xushuang Li
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an 625014, China
| | - Daoshu Luo
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350004, China
| | - Chaobao Zhang
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, China
| | - Shuanggang Hu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, China
| | - Xiangqi Li
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, China
| |
Collapse
|
49
|
Abstract
Fertilization is the process by which eggs and spermatozoa interact, achieve mutual recognition, and fuse to create a zygote, which then develops to form a new individual, thus allowing for the continuity of a species. Despite numerous studies on mammalian fertilization, the molecular mechanisms underpinning the fertilization event remain largely unknown. However, as I summarize here, recent work using both gene-manipulated animals and in vitro studies has begun to elucidate essential sperm and egg molecules and to establish predictive models of successful fertilization.
Collapse
Affiliation(s)
- Masaru Okabe
- Center for Genetic Analysis for Biological Responses Research Institute for Microbial Diseases Osaka University, Yamadaoka 3-1, Suita, Osaka 565-0871, Japan
| |
Collapse
|
50
|
Belleannée C, Calvo É, Caballero J, Sullivan R. Epididymosomes convey different repertoires of microRNAs throughout the bovine epididymis. Biol Reprod 2013; 89:30. [PMID: 23803555 DOI: 10.1095/biolreprod.113.110486] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Epididymosomes are small membrane vesicles that are secreted by epididymal epithelial cells and are involved in posttesticular sperm maturation. Although their role in protein transfer to the sperm membrane is well documented, we report their capacity to transport microRNAs (miRNAs), which are potent regulators of posttranscriptional gene expression. Using a microperfusion technique combined with a global microarray approach, we demonstrated that epididymosomes from two discrete bovine epididymal regions (caput and cauda) possess distinct miRNA signatures. In addition, we also established that miRNA repertoires contained within epididymosomes differ from those of their parent epithelial cells, suggesting that miRNA populations released from the cells may be selectively sorted. Binding of DilC12-labeled epididymosomes to primary cultured epididymal cells was measured by flow cytometry, and the results indicated that epididymosomes from the median caput and their miRNA content may be incorporated into distal caput epithelial cells. Overall, these findings reveal that distinct miRNA repertoires are released into the intraluminal fluid in a region-specific manner and could be involved in a novel mechanism of intercellular communication throughout the epididymis via epididymosomes.
Collapse
Affiliation(s)
- Clémence Belleannée
- CHU de Québec Research Center and Department of Obstetrics and Gynecology, Laval University School of Medicine, Quebec, Canada
| | | | | | | |
Collapse
|