1
|
Chan HY, Robertson SA. Seminal fluid effects on uterine receptivity to embryo implantation: transcriptomic strategies to define molecular mechanisms. Reprod Fertil Dev 2025; 37:RD24162. [PMID: 40100824 DOI: 10.1071/rd24162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
Embryo implantation requires both a developmentally competent embryo and a receptive uterus. Impaired uterine receptivity is a common constraint on implantation success and reproductive outcome. Ovarian steroid hormones oestrogen and progesterone play a central role in establishing uterine receptivity, but other factors also contribute. One additional regulating factor is male partner seminal fluid. However, the full physiological impacts of seminal fluid on uterine receptivity and the specific molecular pathways involved are not yet completely defined. New advances in RNA-sequencing technologies provide a powerful means to examine how uterine tissues and cells respond to seminal fluid contact. Findings utilising sequencing technology provide strong cellular and molecular evidence in humans and mice that seminal fluid contact around the time of ovulation drives immune and vascular changes with potential to affect endometrial receptivity in the peri-implantation phase. This approach has led to the discovery of novel mediators and regulatory factors subsequently shown to facilitate embryo implantation in genetic mouse models, enabling functional validation. Here, we summarise the evidence from recent microarray and RNA-sequencing findings that seminal fluid contact can directly and indirectly impact the transcriptional state of endometrial tissue during the implantation window in mice and also in humans. Progress in elucidating the female reproductive tract response to seminal fluid will improve understanding of male partner effects on endometrial receptivity, and the knowledge gained will have practical applications for achieving healthy pregnancy and offspring outcomes.
Collapse
Affiliation(s)
- Hon Y Chan
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Sarah A Robertson
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
2
|
Xie Y, Xu Z, Wu C, Zhou C, Zhang X, Gu T, Yang J, Yang H, Zheng E, Xu Z, Cai G, Li Z, Liu D, Wu Z, Hong L. Extracellular vesicle-encapsulated miR-21-5p in seminal plasma prevents sperm capacitation via Vinculin inhibition. Theriogenology 2022; 193:103-113. [PMID: 36156422 DOI: 10.1016/j.theriogenology.2022.09.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 07/11/2022] [Accepted: 09/10/2022] [Indexed: 10/31/2022]
Abstract
To penetrate the zona pellucida before sperm-egg binding, sperm must undergo highly time-controlled capacitation and acrosome reaction in the female reproductive tract. Our previous study demonstrated that miR-21-5p is the most abundant miRNA in boar seminal plasma (SP)-derived extracellular vesicles (EVs) and can target Vinculin (VCL) gene, which may participate in boar sperm capacitation. Thus, this study aims to explore the potential role of miR-21-5p from SP-derived EVs in preventing sperm capacitation and its underlying mechanism. We observed that sperm could incorporate miR-21-5p from SP-derived EVs. The roles of SP-derived EVs miR-21-5p in sperm capacitation were then determined using gain- and loss-of-function analyses. In addition, the expression levels of miR-21-5p, VCL, and VCL protein in liquid-preserved boar sperm following transfection were determined using RT-qPCR and Western blotting. Our results revealed that miR-21-5p overexpression inhibited sperm capacitation and acrosome reaction. Similarly, miR-21-5p expression was significantly lower (P < 0.05) in capacitated sperm than un-capacitated sperm. However, the protein level of VCL was also significantly lower (P < 0.05) in capacitated sperm than un-capacitated sperm. Furthermore, immunofluorescence analysis showed that VCL protein mainly located in sperm head and sperm capacitation was inhibited after treating with VCL protein inhibitor (Chrysin). In conclusion, our study provides reasonable evidence that miR-21-5p expression in SP-derived EVs could prevent sperm capacitation via VCL inhibition.
Collapse
Affiliation(s)
- Yanshe Xie
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Zhiqian Xu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Changhua Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Chen Zhou
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | | | - Ting Gu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Jie Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Huaqiang Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Enqin Zheng
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Zheng Xu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Gengyuan Cai
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Zicong Li
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangzhou, 510642, China
| | - Dewu Liu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangzhou, 510642, China.
| | - Linjun Hong
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China.
| |
Collapse
|
3
|
Balu R, Ramachandran SS, Mathimaran A, Jeyaraman J, Paramasivam SG. Functional significance of mouse seminal vesicle sulfhydryl oxidase on sperm capacitation in vitro. Mol Hum Reprod 2022; 29:6637520. [PMID: 35809071 DOI: 10.1093/molehr/gaac025] [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: 01/07/2022] [Revised: 06/06/2022] [Indexed: 11/14/2022] Open
Abstract
During ejaculation, cauda epididymal spermatozoa are suspended in a protein-rich solution of seminal plasma which is composed of proteins mostly secreted from the seminal vesicle. These seminal proteins interact with the sperm cells and bring about changes in their physiology, so that they can become capacitated in order for the fertilization to take place. Sulfhydryl oxidase (SOX) is a member of the QSOX family and its expression is found to be high in the seminal vesicle secretion of mouse. Previously, it has been reported to cross-link thiol containing amino acids among major seminal vesicle secretion (SVS) proteins. However, its role in male reproduction is unclear. In this study, we determined the role of SOX on epididymal sperm maturation and also disclosed the binding effect of SOX on the sperm fertilizing ability in vitro. In order to achieve the above two objectives, we constructed a Sox clone (1.7 kb) using a pET-30a vector. His-tagged recombinant Sox was over expressed in Shuffle Escherichia coli cells and purified using His-Trap column affinity chromatography along with hydrophobic interaction chromatography. The purified SOX was confirmed by Western blot analysis and by its activity with DTT as a substrate. Results obtained from immunocytochemical staining clearly indicated that SOX possesses a binding site on the sperm acrosome. The influence of SOX on oxidation of sperm sulfhydryl to disulfides during epididymal sperm maturation was evaluated by a thiol labelling agent, mBBr. The SOX protein binds on to the sperm cells and increases their progressive motility. The effect of SOX binding on reducing the [Ca2+]i concentration in sperm head, was determined using a calcium probe, Fluo-3 AM. The inhibitory influence of SOX on sperm acrosome reaction was shown by using calcium ionophore A32187 to induce the acrosome reaction. The acrosome-reacted sperm were examined by staining with FITC-conjugated Arachis hypogaea (peanut) lectin. Furthermore, immunocytochemical analysis revealed that SOX remains bound to the sperm cells in the uterus but disappears in the oviduct during their transit in the female reproductive tract. The results from the above experiment revealed that SOX binding on to the sperm acrosome prevents sperm capacitation by affecting the [Ca2+]i concentration in the sperm head and the ionophore-induced acrosome reaction. Thus, the binding of SOX on to the sperm acrosome may possibly serve as a decapacitation factor in the uterus to prevent premature capacitation and acrosome reaction, thus preserving their fertilizing ability.
Collapse
Affiliation(s)
- Rubhadevi Balu
- Department of Biotechnology, BIT-Campus, Anna University, Tiruchirappalli-620024, Tamil Nadu India
| | | | - Amala Mathimaran
- Department of Bioinformatics, Alagappa University, Karaikudi-630 004, Tamil Nadu, India
| | - Jeyakanthan Jeyaraman
- Department of Bioinformatics, Alagappa University, Karaikudi-630 004, Tamil Nadu, India
| | | |
Collapse
|
4
|
Wang TE, Yeh LY, Kuo-Kuang Lee R, Lu CH, Yang TH, Kuo YW, Joshi R, Tsai PS, Li SH. Secretory mouse quiescin sulfhydryl oxidase 1 aggregates defected human and mouse spermatozoa in vitro and in vivo. iScience 2021; 24:103167. [PMID: 34667943 PMCID: PMC8506963 DOI: 10.1016/j.isci.2021.103167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/08/2021] [Accepted: 09/21/2021] [Indexed: 11/25/2022] Open
Abstract
A flavin-dependent enzyme quiescin Q6 sulfhydryl oxidase 1 (QSOX1) catalyzes the oxidation of thiol groups into disulfide bonds. QSOX1 is prominently expressed in the seminal plasma. However, its role in male reproduction is elusive. Here, we purified the secreted form of QSOX1, i.e., QSOX1c, from mouse seminal vesicle secretions and revealed for the first time its function involved in sperm physiology. Exogenous addition of QSOX1c time-dependently promoted the in vitro aggregation of thiol-rich, oxidative stressed, and apoptotic mouse and human sperm cells. Also, in vivo aggregated sperm cells collected from mouse uterine and human ejaculates also showed high levels of QSOX1c, intracellular reactive oxygen species, annexin V, and free thiols. In summary, our studies demonstrated that QSOX1c could agglutinate spermatozoa susceptible to free radical attack and apoptosis. This characteristic may provide an opportunity to separate defective sperm cells and improve sperm quality before artificial insemination in humans and animals. QSOX1c is expressed in the seminal vesicle and presented in the seminal plasma QSOX1c agglutinates thiol-rich, oxidatively stressed, and apoptotic sperm QSOX1c aggregates impaired sperm presented in the mouse uterine and human ejaculates QSOX1c-treated semen may improve the sperm quality for artificial insemination
Collapse
Affiliation(s)
- Tse-En Wang
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, USA
| | - Ling-Yu Yeh
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan
| | - Robert Kuo-Kuang Lee
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan.,Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chung-Hao Lu
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Tsung-Hsien Yang
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan
| | - Yu-Wen Kuo
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Radhika Joshi
- Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Shiue Tsai
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Sheng-Hsiang Li
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan.,MacKay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| |
Collapse
|
5
|
Ou CM, Lee RKK, Lin MH, Lu CH, Yang TH, Yeh LY, Tsai PSJ, Li SH. A mouse seminal vesicle-secreted lysozyme c-like protein modulates sperm capacitation. J Cell Biochem 2021; 122:653-666. [PMID: 33469950 DOI: 10.1002/jcb.29894] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 01/19/2023]
Abstract
Lysozyme (LYZ) c-like proteins are primarily present in the testis and epididymis of male reproductive tissues. Here, we report a novel member of the c-type LYZ family, the seminal vesicle-secreted LYZ c-like protein (SVLLP). Three forms of SVLLP were purified from mouse seminal vesicle secretions and characterized as glycoproteins with the same protein core but different N-linked glycans. SVLLP is structurally similar to c-type LYZ proteins. Only one of the 20 invariant residues was altered in the consensus sequence of c-type LYZs; however, the changed residue (N53S) is one of two essential catalytic residues. LYZ activity assays demonstrated that the three glycoforms of SVLLP lacked enzyme activity. SVLLP is primarily expressed in seminal vesicles. Immunohistochemistry revealed that it occurs in the luminal fluid and mucosal epithelium of the seminal vesicles. Testosterone is not the primary regulator for its expression in the seminal vesicle. SVLLP binds to sperm and suppresses bovine serum albumin-induced sperm capacitation, inhibits the acrosome reaction, and blocks sperm-oocyte interactions in vitro, suggesting that SVLLP is a sperm capacitation inhibitor.
Collapse
Affiliation(s)
- Chung-Mao Ou
- Chemistry Division, Institute of Nuclear Energy Research, Taoyuan City, Taiwan
| | - Robert Kuo-Kuang Lee
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan.,Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ming-Huei Lin
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan.,MacKay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Chung-Hao Lu
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Tsung-Hsien Yang
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan
| | - Ling-Yu Yeh
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan
| | - Pei-Shiue Jason Tsai
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Sheng-Hsiang Li
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan.,MacKay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| |
Collapse
|
6
|
Zalazar L, Stival C, Nicolli AR, De Blas GA, Krapf D, Cesari A. Male Decapacitation Factor SPINK3 Blocks Membrane Hyperpolarization and Calcium Entry in Mouse Sperm. Front Cell Dev Biol 2020; 8:575126. [PMID: 33102481 PMCID: PMC7554638 DOI: 10.3389/fcell.2020.575126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/10/2020] [Indexed: 01/10/2023] Open
Abstract
Mammalian sperm acquire ability to fertilize through a process called capacitation, occurring after ejaculation and regulated by both female molecules and male decapacitation factors. Bicarbonate and calcium present in the female reproductive tract trigger capacitation in sperm, leading to acrosomal responsiveness and hyperactivated motility. Male decapacitating factors present in the semen avert premature capacitation, until detached from the sperm surface. However, their mechanism of action remains elusive. Here we describe for the first time the molecular basis for the decapacitating action of the seminal protein SPINK3 in mouse sperm. When present in the capacitating medium, SPINK3 inhibited Src kinase, a modulator of the potassium channel responsible for plasma membrane hyperpolarization. Lack of hyperpolarization affected calcium channels activity, impairing the acquisition of acrosomal responsiveness and blocking hyperactivation. Interestingly, SPINK3 acted only on non-capacitated sperm, as it did not bind to capacitated cells. Binding selectivity allows its decapacitating action only in non-capacitated sperm, without affecting capacitated cells.
Collapse
Affiliation(s)
- Lucia Zalazar
- Instituto de Investigaciones Biológicas (IIB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | - Cintia Stival
- Laboratory of Cell Signal Transduction Networks, Instituto de Biologia Molecular y Celular de Rosario (IBR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Anabella R Nicolli
- Instituto de Investigaciones Biológicas (IIB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | - Gerardo A De Blas
- Instituto de Histología y Embriología de Mendoza (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, National Scientific and Technical Research Council, Mendoza, Argentina
| | - Dario Krapf
- Laboratory of Cell Signal Transduction Networks, Instituto de Biologia Molecular y Celular de Rosario (IBR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Andreina Cesari
- Instituto de Investigaciones Biológicas (IIB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Mar del Plata, Mar del Plata, Argentina.,Escuela Superior de Medicina, Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| |
Collapse
|
7
|
Anamthathmakula P, Winuthayanon W. Mechanism of semen liquefaction and its potential for a novel non-hormonal contraception†. Biol Reprod 2020; 103:411-426. [PMID: 32529252 PMCID: PMC7523691 DOI: 10.1093/biolre/ioaa075] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 12/21/2022] Open
Abstract
Semen liquefaction is a proteolytic process where a gel-like ejaculated semen becomes watery due to the enzymatic activity of prostate-derived serine proteases in the female reproductive tract. The liquefaction process is crucial for the sperm to gain their motility and successful transport to the fertilization site in Fallopian tubes (or oviducts in animals). Hyperviscous semen or failure in liquefaction is one of the causes of male infertility. Therefore, the biochemical inhibition of serine proteases in the female reproductive tract after ejaculation is a prime target for novel contraceptive development. Herein, we will discuss protein components in the ejaculates responsible for semen liquefaction and any developments of contraceptive methods in the past that involve the liquefaction process.
Collapse
Affiliation(s)
- Prashanth Anamthathmakula
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Wipawee Winuthayanon
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
8
|
Sakaguchi D, Miyado K, Iwamoto T, Okada H, Yoshida K, Kang W, Suzuki M, Yoshida M, Kawano N. Human Semenogelin 1 Promotes Sperm Survival in the Mouse Female Reproductive Tract. Int J Mol Sci 2020; 21:ijms21113961. [PMID: 32486486 PMCID: PMC7312897 DOI: 10.3390/ijms21113961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 01/13/2023] Open
Abstract
Semenogelin 1 (SEMG1), a main component of human seminal plasma, is a multi-functional protein involved in the regulation of sperm motility and fertility. SEMG1 is orthologous to mouse seminal vesicle secretion 2 (SVS2), required for sperm survival in the female reproductive tract after copulation; however, its in vivo function remains unclear. In this study, we addressed this issue by examining the effect of recombinant SEMG1 on intrauterine mouse sperm survival. SEMG1 caused a dose-dependent decrease in mouse sperm motility, similar to its effect on human sperm, but SVS2 had no effect on mouse sperm motility. Mouse epididymal sperm in the presence of 100 µM SEMG1, a concentration that does not affect mouse sperm motility, were injected into the mouse uterus (intrauterine insemination, IUI). IUI combined with SEMG1 significantly increased the survival rate of intrauterine mouse sperm. The effect of SEMG1 on intrauterine sperm survival was comparable with that of SVS2. For clinical applications, three potentially sperm-protecting polypeptides that are easy to handle were designed from SEMG1, but their individual use was unable to mimic the ability of SEMG1. Our results indicate that SEMG1 has potential clinical applications for effective IUI and thereby for safe, simple, and effective internal fertilization.
Collapse
Affiliation(s)
- Daiki Sakaguchi
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan;
| | - Kenji Miyado
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; (K.M.); (W.K.); (M.S.)
| | - Teruaki Iwamoto
- Division of Male Infertility, Center for Human Reproduction, Sanno Hospital, International University of Health and Welfare, Tokyo 107-0052, Japan;
| | - Hiroshi Okada
- Department of Urology, Dokkyo Medical University Saitama Medical Center, Saitama 343-8555, Japan;
| | - Kaoru Yoshida
- Faculty of Biomedical Engineering, Toin University of Yokohama, Kanagawa 225-8503, Japan;
| | - Woojin Kang
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; (K.M.); (W.K.); (M.S.)
| | - Miki Suzuki
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; (K.M.); (W.K.); (M.S.)
| | - Manabu Yoshida
- Misaki Marine Biological Station, School of Science, the University of Tokyo, Kanagawa 238-0225, Japan
- Correspondence: (M.Y.); (N.K.)
| | - Natsuko Kawano
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan;
- Correspondence: (M.Y.); (N.K.)
| |
Collapse
|
9
|
Abstract
Seminal fluid is often assumed to have just one function in mammalian reproduction, delivering sperm to fertilize oocytes. But seminal fluid also transmits signaling agents that interact with female reproductive tissues to facilitate conception and .pregnancy. Upon seminal fluid contact, female tissues initiate a controlled inflammatory response that affects several aspects of reproductive function to ultimately maximize the chances of a male producing healthy offspring. This effect is best characterized in mice, where the female response involves several steps. Initially, seminal fluid factors cause leukocytes to infiltrate the female reproductive tract, and to selectively target and eliminate excess sperm. Other signals stimulate ovulation, induce an altered transcriptional program in female tract tissues that modulates embryo developmental programming, and initiate immune adaptations to promote receptivity to implantation and placental development. A key result is expansion of the pool of regulatory T cells that assist implantation by suppressing inflammation, mediating tolerance to male transplantation antigens, and promoting uterine vascular adaptation and placental development. Principal signaling agents in seminal fluid include prostaglandins and transforming growth factor-β. The balance of male signals affects the nature of the female response, providing a mechanism of ‟cryptic female choiceˮ that influences female reproductive investment. Male-female seminal fluid signaling is evident in all mammalian species investigated including human, and effects of seminal fluid in invertebrates indicate evolutionarily conserved mechanisms. Understanding the female response to seminal fluid will shed new light on infertility and pregnancy disorders and is critical to defining how events at conception influence offspring health.
Collapse
Affiliation(s)
- John E Schjenken
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
| |
Collapse
|
10
|
Deletion of a Seminal Gene Cluster Reinforces a Crucial Role of SVS2 in Male Fertility. Int J Mol Sci 2019; 20:ijms20184557. [PMID: 31540031 PMCID: PMC6769479 DOI: 10.3390/ijms20184557] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 11/17/2022] Open
Abstract
Multiple genes, whose functions or expression are overlapping, compensate for the loss of one gene. A gene cluster in the mouse genome encodes five seminal vesicle proteins (SVS2, SVS3, SVS4, SVS5, and SVS6). These proteins are produced by male rodents and function in formation of the copulatory plug following mating. SVS2 plays an essential role in the successful internal fertilization by protecting the sperm membrane against a uterine immune attack. We hypothesized that the four remaining seminal vesicle proteins (SVPs) of this gene cluster may partially/completely compensate for the deficiency of SVS2. For confirming our hypothesis, we generated mice lacking the entire SVP-encoding gene cluster and compared their fecundity with Svs2-deficient (Svs2−/−) mice; that is, mice deficient in Svs2 alone. A single loxP site remained after the deletion of the Svs2 gene. Therefore, we inserted another loxP site by combining the CRISPR/Cas9 system with single-stranded oligodeoxynucleotides (ssODN). Male mice lacking the entire SVP-encoding gene cluster (Svs2–6−/− mice) and thereby all five SVP proteins, generated by the deletion of 100kbp genomic DNA, showed low fecundity. However, the fecundity level was comparable with that from Svs2−/− male mice. Our results demonstrate that SVS3, SVS4, SVS5, and SVS6 do not function in the protection of sperm against a uterine immune attack in the absence of SVS2. Thus, Svs2 is the critical gene in the SVP gene cluster.
Collapse
|
11
|
Civetta A, Ranz JM. Genetic Factors Influencing Sperm Competition. Front Genet 2019; 10:820. [PMID: 31572439 PMCID: PMC6753916 DOI: 10.3389/fgene.2019.00820] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/08/2019] [Indexed: 12/26/2022] Open
Abstract
Females of many different species often mate with multiple males, creating opportunities for competition among their sperm. Although originally unappreciated, sperm competition is now considered a central form of post-copulatory male–male competition that biases fertilization. Assays of differences in sperm competitive ability between males, and interactions between females and males, have made it possible to infer some of the main mechanisms of sperm competition. Nevertheless, classical genetic approaches have encountered difficulties in identifying loci influencing sperm competitiveness while functional and comparative genomic methodologies, as well as genetic variant association studies, have uncovered some interesting candidate genes. We highlight how the systematic implementation of approaches that incorporate gene perturbation assays in experimental competitive settings, together with the monitoring of progeny output or sperm features and behavior, has allowed the identification of genes unambiguously linked to sperm competitiveness. The emerging portrait from 45 genes (33 from fruit flies, 8 from rodents, 2 from nematodes, and 2 from ants) is their remarkable breadth of biological roles exerted through males and females, the non-preponderance of sperm genes, and their overall pleiotropic nature.
Collapse
Affiliation(s)
- Alberto Civetta
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
| | - José M Ranz
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA, United States
| |
Collapse
|
12
|
Genetic resistance to DEHP-induced transgenerational endocrine disruption. PLoS One 2019; 14:e0208371. [PMID: 31181066 PMCID: PMC6557477 DOI: 10.1371/journal.pone.0208371] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/15/2019] [Indexed: 02/07/2023] Open
Abstract
Di(2-ethylhexyl)phthalate (DEHP) interferes with sex hormones signaling pathways (SHP). C57BL/6J mice prenatally exposed to 300 mg/kg/day DEHP develop a testicular dysgenesis syndrome (TDS) at adulthood, but similarly-exposed FVB/N mice are not affected. Here we aim to understand the reasons behind this drastic difference that should depend on the genome of the strain. In both backgrounds, pregnant female mice received per os either DEHP or corn oil vehicle and the male filiations were examined. Computer-assisted sperm analysis showed a DEHP-induced decreased sperm count and velocities in C57BL/6J. Sperm RNA sequencing experiments resulted in the identification of the 62 most differentially expressed RNAs. These RNAs, mainly regulated by hormones, produced strain-specific transcriptional responses to prenatal exposure to DEHP; a pool of RNAs was increased in FVB, another pool of RNAs was decreased in C57BL/6J. In FVB/N, analysis of non-synonymous single nucleotide polymorphisms (SNP) impacting SHP identified rs387782768 and rs29315913 respectively associated with absence of the Forkhead Box A3 (Foxa3) RNA and increased expression of estrogen receptor 1 variant 4 (NM_001302533) RNA. Analysis of the role of SNPs modifying SHP binding sites in function of strain-specific responses to DEHP revealed a DEHP-resistance allele in FVB/N containing an additional FOXA1-3 binding site at rs30973633 and four DEHP-induced beta-defensins (Defb42, Defb30, Defb47 and Defb48). A DEHP-susceptibility allele in C57BL/6J contained five SNPs (rs28279710, rs32977910, rs46648903, rs46677594 and rs48287999) affecting SHP and six genes (Svs2, Svs3b, Svs4, Svs3a, Svs6 and Svs5) epigenetically silenced by DEHP. Finally, targeted experiments confirmed increased methylation in the Svs3ab promoter with decreased SEMG2 persisting across generations, providing a molecular explanation for the transgenerational sperm velocity decrease found in C57BL/6J after DEHP exposure. We conclude that the existence of SNP-dependent mechanisms in FVB/N inbred mice may confer resistance to transgenerational endocrine disruption.
Collapse
|
13
|
Xiong W, Wang Z, Shen C. An update of the regulatory factors of sperm migration from the uterus into the oviduct by genetically manipulated mice. Mol Reprod Dev 2019; 86:935-955. [PMID: 31131960 DOI: 10.1002/mrd.23180] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/04/2019] [Accepted: 05/09/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Wenfeng Xiong
- State Key Laboratory of Medical Genomics, Research Center for Experimental MedicineShanghai Rui‐Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
- School of Life Sciences and BiochemistryShanghai Jiao Tong University Shanghai China
| | - Zhugang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental MedicineShanghai Rui‐Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Chunling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental MedicineShanghai Rui‐Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
14
|
Arai Y, Sakase M, Fukushima M, Harayama H. Identification of isoforms of calyculin A-sensitive protein phosphatases which suppress full-type hyperactivation in bull ejaculated spermatozoa. Theriogenology 2019; 129:46-53. [PMID: 30798082 DOI: 10.1016/j.theriogenology.2019.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/04/2019] [Accepted: 02/10/2019] [Indexed: 01/04/2023]
Abstract
In bull spermatozoa, extracellular Ca2+-dependent full-type hyperactivation, which is characterized by the asymmetrical beating in whole parts of the middle/principal pieces, is suppressed by calyculin A-sensitive protein phosphatases. The aim of this study was to identify isoforms of these protein phosphatases. Ejaculated spermatozoa were used for the investigation on effects of protein phosphatase inhibitors (calyculin A with high specificity for both of protein phosphatases 1 and 2A, and okadaic acid with relatively higher specificity for protein phosphatase 2A than protein phosphatase 1) on the induction of extracellular Ca2+-dependent full-type hyperactivation by incubation with CaCl2 and cAMP analog (cBiMPS). They were also used for the immunodetection of protein phosphatases 1α, 1β, 1γ, 2Aα and 2Aβ. Percentages of full-type hyperactivated spermatozoa significantly increased after incubation with calyculin A (10 nM) in a concentration-dependent manner of CaCl2 (0-3.42 mM), though only minor increases in the percentages of full-type hyperactivated spermatozoa were observed after incubation with okadaic acid (10 nM). Moreover, the immunodetection of protein phosphatase isoforms showed sperm connecting piece and flagellum included protein phosphatases 1α and 1γ, but did not do the other isoforms. These results suggest that calyculin A-sensitive and okadaic acid-less sensitive protein phosphatases (1α and 1γ) are suppressors for the extracellular Ca2+-dependent full-type hyperactivation in bull ejaculated spermatozoa.
Collapse
Affiliation(s)
- Yuka Arai
- Laboratory of Reproductive Biology, Division of Animal Science, Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Japan
| | - Mitsuhiro Sakase
- Hokubu Agricultural Institute, Hyogo Prefectural Technology Center for Agriculture, Forestry and Fisheries, Japan
| | - Moriyuki Fukushima
- Hokubu Agricultural Institute, Hyogo Prefectural Technology Center for Agriculture, Forestry and Fisheries, Japan
| | - Hiroshi Harayama
- Laboratory of Reproductive Biology, Division of Animal Science, Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Japan.
| |
Collapse
|
15
|
Yu H, Hackenbroch L, Meyer FRL, Reiser J, Razzazi-Fazeli E, Nöbauer K, Besenfelder U, Vogl C, Brem G, Mayrhofer C. Identification of Rabbit Oviductal Fluid Proteins Involved in Pre-Fertilization Processes by Quantitative Proteomics. Proteomics 2019; 19:e1800319. [PMID: 30637940 DOI: 10.1002/pmic.201800319] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 12/28/2018] [Indexed: 01/28/2023]
Abstract
Oviductal fluid (ODF) proteins modulate and support reproductive processes in the oviduct. In the present study, proteins involved in the biological events that precede fertilization have been identified in the rabbit ODF proteome, isolated from the ampulla and isthmus of the oviduct at different time points within 8 h after intrauterine insemination. A workflow is used that integrates lectin affinity capture with stable-isotope dimethyl labeling prior to nanoLC-MS/MS analysis. In total, over 400 ODF proteins, including 214 lectin enriched glycoproteins, are identified and quantified. Selected data are validated by Western blot analysis. Spatiotemporal alterations in the abundance of ODF proteins in response to insemination are detected by global analysis. A subset of 63 potentially biologically relevant ODF proteins is identified, including extracellular matrix components, chaperones, oxidoreductases, and immunity proteins. Functional enrichment analysis reveals an altered peptidase regulator activity upon insemination. In addition to protein identification and abundance changes, N-glycopeptide analysis further identifies 281 glycosites on 199 proteins. Taken together, these results show, for the first time, the evolving oviductal milieu early upon insemination. The identified proteins are likely those that modulate in vitro processes, including spermatozoa function.
Collapse
Affiliation(s)
- Hans Yu
- Institute of Biotechnology in Animal Production, Interuniversity Department for Agrobiotechnology (IFA Tulln), University of Natural Resources and Applied Life Sciences Vienna, 3430, Tulln, Austria.,Institute of Animal Breeding and Genetics, Department for Biomedical Sciences, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Lena Hackenbroch
- Institute of Biotechnology in Animal Production, Interuniversity Department for Agrobiotechnology (IFA Tulln), University of Natural Resources and Applied Life Sciences Vienna, 3430, Tulln, Austria.,Institute of Animal Breeding and Genetics, Department for Biomedical Sciences, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Florian R L Meyer
- Institute of Biotechnology in Animal Production, Interuniversity Department for Agrobiotechnology (IFA Tulln), University of Natural Resources and Applied Life Sciences Vienna, 3430, Tulln, Austria.,Institute of Animal Breeding and Genetics, Department for Biomedical Sciences, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Judith Reiser
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilian University, 85764, Munich, Germany
| | - Ebrahim Razzazi-Fazeli
- VetCore Facility for Research, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Katharina Nöbauer
- VetCore Facility for Research, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Urban Besenfelder
- Reproduction Centre Wieselburg, University of Veterinary Medicine Vienna, 3250, Vienna, Austria
| | - Claus Vogl
- Institute of Animal Breeding and Genetics, Department for Biomedical Sciences, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Gottfried Brem
- Institute of Biotechnology in Animal Production, Interuniversity Department for Agrobiotechnology (IFA Tulln), University of Natural Resources and Applied Life Sciences Vienna, 3430, Tulln, Austria.,Institute of Animal Breeding and Genetics, Department for Biomedical Sciences, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Corina Mayrhofer
- Institute of Biotechnology in Animal Production, Interuniversity Department for Agrobiotechnology (IFA Tulln), University of Natural Resources and Applied Life Sciences Vienna, 3430, Tulln, Austria.,Institute of Animal Breeding and Genetics, Department for Biomedical Sciences, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| |
Collapse
|
16
|
Harayama H. Flagellar hyperactivation of bull and boar spermatozoa. Reprod Med Biol 2018; 17:442-448. [PMID: 30377397 PMCID: PMC6194283 DOI: 10.1002/rmb2.12227] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In mammals, flagellar hyperactivation is indispensable to sperm fertilization with oocytes in vivo, although there are species differences in regulatory mechanisms for this event. In this study, I reviewed researches regarding hyperactivation of bull and boar spermatozoa, in comparison with those of spermatozoa from other species. METHODS Recent publications regarding sperm hyperactivation were collected and summarized. RESULTS MAIN FINDINGS In bull and boar spermatozoa, there are two types of hyperactivation "full-type hyperactivation and nonfull-type hyperactivation" which are equivalent to anti-hock hyperactivation and pro-hock hyperactivation of mouse spermatozoa, respectively, on the basis of the flagellar parts exhibiting asymmetrical beating. Full-type hyperactivation is initiated in response to a rapid increase of cytoplasmic Ca2+ in the connecting/middle and principal pieces by the mobilization of this divalent ion from extracellular space and internal store through cation channels. Regulatory molecules for the increase of cytoplasmic Ca2+ in the connecting/middle pieces are probably different from those in the principal pieces. CONCLUSION I have proposed a hypothesis on the regulation of full-type hyperactivation by the distinct signaling cascades leading to the increase in cytoplasmic Ca2+ between the connecting/middle and principal pieces of bull and boar spermatozoa.
Collapse
Affiliation(s)
- Hiroshi Harayama
- Division of Animal Science, Department of Bioresource Science, Graduate School of Agricultural ScienceKobe UniversityKobeJapan
| |
Collapse
|
17
|
Li SH, Hwu YM, Lu CH, Lin MH, Yeh LY, Lee RKK. Serine Protease Inhibitor SERPINE2 Reversibly Modulates Murine Sperm Capacitation. Int J Mol Sci 2018; 19:ijms19051520. [PMID: 29783741 PMCID: PMC5983788 DOI: 10.3390/ijms19051520] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/17/2018] [Accepted: 05/17/2018] [Indexed: 12/16/2022] Open
Abstract
SERPINE2 (serpin peptidase inhibitor, clade E, member 2), predominantly expressed in the seminal vesicle, can inhibit murine sperm capacitation, suggesting its role as a sperm decapacitation factor (DF). A characteristic of DF is its ability to reverse the capacitation process. Here, we investigated whether SERPINE2 can reversibly modulate sperm capacitation. Immunocytochemical staining revealed that SERPINE2 was bound onto both capacitated and uncapacitated sperm. It reversed the increase in BSA-induced sperm protein tyrosine phosphorylation levels. The effective dose and incubation time were found to be >0.1 mg/mL and >60 min, respectively. Calcium ion levels in the capacitated sperm were reduced to a level similar to that in uncapacitated sperm after 90 min of incubation with SERPINE2. In addition, the acrosome reaction of capacitated sperm was inhibited after 90 min of incubation with SERPINE2. Oviductal sperm was readily induced to undergo the acrosome reaction using the A23187 ionophore; however, the acrosome reaction was significantly reduced after incubation with SERPINE2 for 60 and 120 min. These findings suggested that SERPINE2 prevented as well as reversed sperm capacitation in vitro. It also prevented the acrosome reaction in in vivo-capacitated sperm isolated from the oviduct. Thus, SERPINE2 could reversibly modulate murine sperm capacitation.
Collapse
Affiliation(s)
- Sheng-Hsiang Li
- Department of Medical Research, Mackay Memorial Hospital, Tamsui District, New Taipei City 251, Taiwan.
- Mackay Junior College of Medicine, Nursing, and Management, Beitou District, Taipei City 112, Taiwan.
| | - Yuh-Ming Hwu
- Department of Medical Research, Mackay Memorial Hospital, Tamsui District, New Taipei City 251, Taiwan.
- Mackay Junior College of Medicine, Nursing, and Management, Beitou District, Taipei City 112, Taiwan.
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei City 104, Taiwan.
- Mackay Medical College, Sanzhi District, New Taipei City 252, Taiwan.
| | - Chung-Hao Lu
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei City 104, Taiwan.
| | - Ming-Huei Lin
- Mackay Junior College of Medicine, Nursing, and Management, Beitou District, Taipei City 112, Taiwan.
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei City 104, Taiwan.
| | - Ling-Yu Yeh
- Department of Medical Research, Mackay Memorial Hospital, Tamsui District, New Taipei City 251, Taiwan.
| | - Robert Kuo-Kuang Lee
- Department of Medical Research, Mackay Memorial Hospital, Tamsui District, New Taipei City 251, Taiwan.
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei City 104, Taiwan.
- Department of Obstetrics and Gynecology, Taipei Medical University, Taipei City 110, Taiwan.
| |
Collapse
|
18
|
Fujihara Y, Miyata H, Ikawa M. Factors controlling sperm migration through the oviduct revealed by gene-modified mouse models. Exp Anim 2018; 67:91-104. [PMID: 29353867 PMCID: PMC5955741 DOI: 10.1538/expanim.17-0153] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mammalian fertilization is comprised of many steps including sperm survival in the
uterus, sperm migration in the female reproductive tract, physiological and morphological
changes to the spermatozoa, and sperm-egg interaction in the oviduct. In
vitro studies have revealed essential factors for these fertilization steps for
over half a century. However, the molecular mechanism of fertilization has recently been
revised by the emergence of genetically modified animals. Here, we focus on essential
factors for sperm fertilizing ability and describe recent advances in our knowledge of the
mechanisms of mammalian fertilization, especially of sperm migration from the uterus into
the oviduct.
Collapse
Affiliation(s)
- Yoshitaka Fujihara
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan.,The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
19
|
Yamasaki K, Yoshida K, Yoshiike M, Shimada K, Nishiyama H, Takamizawa S, Yanagida K, Iwamoto T. Relationship between Semenogelins bound to human sperm and other semen parameters and pregnancy outcomes. Basic Clin Androl 2017; 27:15. [PMID: 28794880 PMCID: PMC5547539 DOI: 10.1186/s12610-017-0059-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/24/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Semenogelins (SEMGs) are major components of human seminal vesicle secretions. Due to SEMG's sperm-motility inhibitor, a significant negative correlation between sperm motility and the proportion of SEMG-bound spermatozoa (SEMG+) was found in asthenozoospermic patients. SEMGs also show intrinsic inhibitory capability for sperm capacitation; however, studies on actual clinical specimens have not been conducted. METHODS To reveal the relationship between SEMGs and the fertilizing capacity of sperm from male infertile patients who are not restricted to asthenozoospermia, we measured the proportion of SEMG+ in the spermatozoa of 142 male infertile patients. The pregnancy outcomes in partners of these patients were retrospectively analyzed using questionnaires. RESULTS Among examined semen parameters, only the total SEMG-unbound sperm count showed a tendency to be different between the spontaneous pregnancy or intra-uterine-insemination-pregnancy groups and in-vitro-fertilization- or intracytoplasmic-sperm-injection-pregnancy groups. It was elevated in the former group, which includes patients who used in vivo fertilization. CONCLUSIONS The total SEMG-unbound sperm count would be a relevant parameter for in vivo fertilization. This result suggests that SEMGs inhibit ectopic capacitation before sperm reach the fertilization site and that the number of total SEMG-unbound sperm is a parameter directly linked to the possibility of in vivo fertilization.
Collapse
Affiliation(s)
- Kazumitsu Yamasaki
- Department of Urology, Tsukuba Gakuen Hospital, Tsukuba, Japan
- Center for IVF and Infertility, International University of Health and Welfare Hospital, Nasushiobara, Japan
| | - Kaoru Yoshida
- Faculty of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Miki Yoshiike
- Department of Urology, St Marianna University School of Medicine, Kawasaki, Japan
| | - Kazuhiko Shimada
- Institute for Central Clinic, Shimotsuke, Japan
- Center for IVF and Infertility, International University of Health and Welfare Hospital, Nasushiobara, Japan
| | - Hiroyuki Nishiyama
- Department of Urology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoru Takamizawa
- Center for IVF and Infertility, International University of Health and Welfare Hospital, Nasushiobara, Japan
| | - Kaoru Yanagida
- Center for IVF and Infertility, International University of Health and Welfare Hospital, Nasushiobara, Japan
| | - Teruaki Iwamoto
- Center for IVF and Infertility, International University of Health and Welfare Hospital, Nasushiobara, Japan
| |
Collapse
|
20
|
Testicular Dysgenesis Syndrome and Long-Lasting Epigenetic Silencing of Mouse Sperm Genes Involved in the Reproductive System after Prenatal Exposure to DEHP. PLoS One 2017; 12:e0170441. [PMID: 28085963 PMCID: PMC5234833 DOI: 10.1371/journal.pone.0170441] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/04/2017] [Indexed: 11/19/2022] Open
Abstract
The endocrine disruptor bis(2-ethylhexyl) phthalate (DEHP) has been shown to exert adverse effects on the male animal reproductive system. However, its mode of action is unclear and a systematic analysis of its molecular targets is needed. In the present study, we investigated the effects of prenatal exposure to 300 mg/kg/day DEHP during a critical period for gonads differentiation to testes on male mice offspring reproductive parameters, including the genome-wide RNA expression and associated promoter methylation status in the sperm of the first filial generation. It was observed that adult male offspring displayed symptoms similar to the human testicular dysgenesis syndrome. A combination of sperm transcriptome and methylome data analysis allowed to detect a long-lasting DEHP-induced and robust promoter methylation-associated silencing of almost the entire cluster of the seminal vesicle secretory proteins and antigen genes, which are known to play a fundamental role in sperm physiology. It also resulted in the detection of a DEHP-induced promoter demethylation associated with an up-regulation of three genes apparently not relevant for sperm physiology and partially related to the immune system. As previously reported, DEHP induced an increase in mir-615 microRNA expression and a genome-wide decrease in microRNA promoter methylation. A functional analysis revealed DEHP-induced enrichments in down-regulated gene transcripts coding for peroxisome proliferator-activated receptors and tumor necrosis factor signaling pathways, and in up-regulated gene transcripts coding for calcium binding and numerous myosin proteins. All these enriched pathways and networks have been described to be associated in some way with the reproductive system. This study identifies a large new array of genes dysregulated by DEHP that may play a role in the complex system controlling the development of the male reproductive system.
Collapse
|
21
|
Araki N, Kawano N, Kang W, Miyado K, Yoshida K, Yoshida M. Seminal vesicle proteins SVS3 and SVS4 facilitate SVS2 effect on sperm capacitation. Reproduction 2016; 152:313-21. [PMID: 27486266 DOI: 10.1530/rep-15-0551] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 08/01/2016] [Indexed: 12/21/2022]
Abstract
Mammalian spermatozoa acquire their fertilizing ability in the female reproductive tract (sperm capacitation). On the other hand, seminal vesicle secretion, which is a major component of seminal plasma, inhibits the initiation of sperm capacitation (capacitation inhibition) and reduces the fertility of the capacitated spermatozoa (decapacitation). There are seven major proteins involved in murine seminal vesicle secretion (SVS1-7), and we have previously shown that SVS2 acts as both a capacitation inhibitor and a decapacitation factor, and is indispensable for in vivo fertilization. However, the effects of SVSs other than SVS2 on the sperm have not been elucidated. Since mouse Svs2-Svs6 genes evolved by gene duplication belong to the same gene family, it is possible that SVSs other than SVS2 also have some effects on sperm capacitation. In this study, we examined the effects of SVS3 and SVS4 on sperm capacitation. Our results showed that both SVS3 and SVS4 are able to bind to spermatozoa, but SVS3 alone showed no effects on sperm capacitation. On the other hand, SVS4 acted as a capacitation inhibitor, although it did not show decapacitation abilities. Interestingly, SVS3 showed an affinity for SVS2 and it facilitated the effects of SVS2. Interaction of SVS2 and spermatozoa is mediated by the ganglioside GM1 in the sperm membrane; however, both SVS3 and SVS4 had weaker affinities for GM1 than SVS2. Therefore, we suggest that separate processes may cause capacitation inhibition and decapacitation, and SVS3 and SVS4 act on sperm capacitation cooperatively with SVS2.
Collapse
Affiliation(s)
- Naoya Araki
- Misaki Marine Biological StationSchool of Science, The University of Tokyo, Miura, Kanagawa, Japan
| | - Natsuko Kawano
- Department of AgricultureMeiji University, Kawasaki, Kanagawa, Japan Department of Reproductive BiologyNational Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Woojin Kang
- Department of AgricultureMeiji University, Kawasaki, Kanagawa, Japan Department of Reproductive BiologyNational Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Kenji Miyado
- Department of Reproductive BiologyNational Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Kaoru Yoshida
- Biomedical Engineering CenterToin University of Yokohama, Yokohama, Kanagawa, Japan
| | - Manabu Yoshida
- Misaki Marine Biological StationSchool of Science, The University of Tokyo, Miura, Kanagawa, Japan Center for Marine BiologyThe University of Tokyo, Miura, Kanagawa, Japan
| |
Collapse
|