Juno is the egg Izumo receptor and is essential for mammalian fertilization

Enrica Bianchi, Brendan Doe, David Goulding, Gavin J Wright, Enrica Bianchi, Brendan Doe, David Goulding, Gavin J Wright

Abstract

Fertilization occurs when sperm and egg recognize each other and fuse to form a new, genetically distinct organism. The molecular basis of sperm-egg recognition is unknown, but is likely to require interactions between receptor proteins displayed on their surface. Izumo1 is an essential sperm cell-surface protein, but its receptor on the egg has not been described. Here we identify folate receptor 4 (Folr4) as the receptor for Izumo1 on the mouse egg, and propose to rename it Juno. We show that the Izumo1-Juno interaction is conserved within several mammalian species, including humans. Female mice lacking Juno are infertile and Juno-deficient eggs do not fuse with normal sperm. Rapid shedding of Juno from the oolemma after fertilization suggests a mechanism for the membrane block to polyspermy, ensuring eggs normally fuse with just a single sperm. Our discovery of an essential receptor pair at the nexus of conception provides opportunities for the rational development of new fertility treatments and contraceptives.

Figures

Figure 1. Juno is the GPI-anchored oocyte…
Figure 1. Juno is the GPI-anchored oocyte surface receptor for Izumo1
(a) An avid recombinant Izumo1 protein but not a control bound the oolemma. (b) Izumo1 bound HEK293 cells transfected with a Juno cDNA (clone B2) but not untransfected controls. (c) Juno is highly expressed on the oolemma of unfertilised eggs. (d) Preincubation of eggs with an anti-Juno but not an anti-Cd55 control antibody blocked binding of recombinant Izumo1 protein. (e, f) Cell surface Juno was lost after treatment with PIPLC on Juno-transfected HEK293 cells (e) or eggs (f). Images are single optical sections of unfertilised mouse metaphase II eggs; scale bars are 20 μm in a, c, d, f, and 10 μm in b.
Figure 2. The Juno-Izumo1 interaction is direct,…
Figure 2. The Juno-Izumo1 interaction is direct, transient and conserved across mammals
(a) Biophysical analysis of the Juno-Izumo1 interaction using SPR. Serial dilutions of purified, soluble Juno were injected over immobilised Izumo1, and kinetic parameters derived from a 1:1 Langmuir binding model (red lines). (b) Binding specificity within the paralogues of the mouse Izumo and Folr families: only Juno (Folr4) bound Izumo1. (c) The Juno-Izumo1 interaction is conserved across mammals. AVEXIS was used for binding analysis in (b) and (c) using recombinant Folr/Juno proteins as preys and Izumo proteins as baits; control bait in (c) was Cd4. All bar charts show mean ± SEM; n =3.
Figure 3. Juno is essential for female…
Figure 3. Juno is essential for female fertility
(a) An anti-Juno monoclonal antibody potently blocked in-vitro fertilisation; anti-Izumo1 is shown for comparison. (b) Female Juno−/− mice are infertile. Continuous mating of Juno−/− female mice to proven wild-type males for three months did not result in any pups. (c) Greater numbers of sperm were observed in the perivitelline space (PVS) of eggs from superovulated Juno-deficient (−/−) mice relative to wild-type (+/+). (d, e) Juno−/− eggs do not fuse with wild-type sperm in vitro. Sperm were added to Juno-deficient (−/−) and wild-type (+/+) eggs and fused sperm quantified after two hours (d), or pronuclei after six hours (e). All bar charts show mean ± SEM; numbers in parentheses are total number of eggs (a, c, d, e) or number of mating pairs (b).
Figure 4. Normally-fertilised, but not ICSI-fertilised or…
Figure 4. Normally-fertilised, but not ICSI-fertilised or parthenogenetically-activated eggs, rapidly shed Juno from the oolemma
(a) Cell surface Juno rapidly becomes undetectable after fertilisation. Juno (green) is expressed on ovulated metaphase II eggs but is barely evident at telophase II and undetectable on pronuclear-stage zygotes. Arrow and asterisk indicate sites of first and second polar body extrusion respectively; chromosomes are not within the plane of focus. (b) Immunogold electron microscopy localised Juno primarily to the oolemma in unfertilised, ovulated eggs but was redistributed to vesicles within the perivitelline space within one hour after fertilisation. (c) Eggs fertilised by ICSI or parthenogenetically-activated retain oolemmal Juno staining until at least the pronuclear stage, as shown. Nuclei/pronuclei were visualised with DAPI (blue). Images show representative eggs from three independent experiments. Scale bars represent 20 μm (a and c) and 1 μm (b).

References

    1. Okabe M. The cell biology of mammalian fertilization. Development. 2013;140:4471–4479.
    1. Gardner AJ, Evans JP. Mammalian membrane block to polyspermy: new insights into how mammalian eggs prevent fertilisation by multiple sperm. Reprod Fertil Dev. 2006;18:53–61.
    1. Ikawa M, Inoue N, Benham AM, Okabe M. Fertilization: a sperm’s journey to and interaction with the oocyte. J Clin Invest. 2010;120:984–994.
    1. Inoue N, Ikawa M, Isotani A, Okabe M. The immunoglobulin superfamily protein Izumo is required for sperm to fuse with eggs. Nature. 2005;434:234–238.
    1. Le Naour F, Rubinstein E, Jasmin C, Prenant M, Boucheix C. Severely reduced female fertility in CD9-deficient mice. Science. 2000;287:319–321.
    1. Miyado K, et al. Requirement of CD9 on the egg plasma membrane for fertilization. Science. 2000;287:321–324.
    1. Kaji K, et al. The gamete fusion process is defective in eggs of Cd9-deficient mice. Nat Genet. 2000;24:279–282.
    1. Satouh Y, Inoue N, Ikawa M, Okabe M. Visualization of the moment of mouse sperm-egg fusion and dynamic localization of IZUMO1. J Cell Sci. 2012;125:4985–4990.
    1. Inoue N, et al. Molecular dissection of IZUMO1, a sperm protein essential for sperm-egg fusion. Development. 2013;140:3221–3229.
    1. Coonrod SA, et al. Treatment of mouse oocytes with PI-PLC releases 70-kDa (pI 5) and 35- to 45-kDa (pI 5.5) protein clusters from the egg surface and inhibits sperm-oolemma binding and fusion. Dev Biol. 1999;207:334–349.
    1. Alfieri JA, et al. Infertility in female mice with an oocyte-specific knockout of GPI-anchored proteins. J Cell Sci. 2003;116:2149–2155.
    1. Wright GJ. Signal initiation in biological systems: the properties and detection of transient extracellular protein interactions. Mol Biosyst. 2009;5:1405–1412.
    1. Yamaguchi T, et al. Control of immune responses by antigen-specific regulatory T cells expressing the folate receptor. Immunity. 2007;27:145–159.
    1. Teng MW, et al. Multiple antitumor mechanisms downstream of prophylactic regulatory T-cell depletion. Cancer Res. 2010;70:2665–2674.
    1. Liang SC, Moskalenko M, Van Roey M, Jooss K. Depletion of regulatory T cells by targeting folate receptor 4 enhances the potency of a GM-CSF-secreting tumor cell immunotherapy. Clin Immunol. 2013;148:287–298.
    1. Kunisawa J, Hashimoto E, Ishikawa I, Kiyono H. A pivotal role of vitamin B9 in the maintenance of regulatory T cells in vitro and in vivo. PLoS One. 2012;7:e32094.
    1. Kinoshita M, et al. Dietary folic acid promotes survival of Foxp3+ regulatory T cells in the colon. J Immunol. 2012;189:2869–2878.
    1. Salbaum JM, Kruger C, Kappen C. Mutation at the folate receptor 4 locus modulates gene expression profiles in the mouse uterus in response to periconceptional folate supplementation. Biochim Biophys Acta. 2013;1832:1653–1661.
    1. Chen C, et al. Structural basis for molecular recognition of folic acid by folate receptors. Nature. 2013;500:486–489.
    1. Wibowo AS, et al. Structures of human folate receptors reveal biological trafficking states and diversity in folate and antifolate recognition. Proc Natl Acad Sci U S A. 2013;110:15180–15188.
    1. Jia Z, et al. A novel splice variant of FR4 predominantly expressed in CD4+CD25+ regulatory T cells. Immunol Invest. 2009;38:718–729.
    1. Ellerman DA, et al. Izumo is part of a multiprotein family whose members form large complexes on mammalian sperm. Mol Reprod Dev. 2009;76:1188–1199.
    1. Bushell KM, Sollner C, Schuster-Boeckler B, Bateman A, Wright GJ. Large-scale screening for novel low-affinity extracellular protein interactions. Genome Res. 2008;18:622–630.
    1. Jaffe LA. Fast block to polyspermy in sea urchin eggs is electrically mediated. Nature. 1976;261:68–71.
    1. Sabharanjak S, Mayor S. Folate receptor endocytosis and trafficking. Adv Drug Deliv Rev. 2004;56:1099–1109.
    1. Jackowski S, Dumont JN. Surface alterations of the mouse zona pellucida and ovum following in vivo fertilization: correlation with the cell cycle. Biol Reprod. 1979;20:150–161.
    1. Podbilewicz B, et al. The C. elegans developmental fusogen EFF-1 mediates homotypic fusion in heterologous cells and in vivo. Dev Cell. 2006;11:471–481.
    1. Mi S, et al. Syncytin is a captive retroviral envelope protein involved in human placental morphogenesis. Nature. 2000;403:785–789.
    1. Abmayr SM, Pavlath GK. Myoblast fusion: lessons from flies and mice. Development. 2012;139:641–656.
    1. Powell GT, Wright GJ. Jamb and jamc are essential for vertebrate myocyte fusion. PLoS Biol. 2011;9:e1001216.
    1. Millay DP, et al. Myomaker is a membrane activator of myoblast fusion and muscle formation. Nature. 2013;499:301–305.
    1. Hemler ME. Tetraspanin functions and associated microdomains. Nat Rev Mol Cell Biol. 2005;6:801–811.
    1. Jegou A, et al. CD9 tetraspanin generates fusion competent sites on the egg membrane for mammalian fertilization. Proc Natl Acad Sci U S A. 2011;108:10946–10951.
    1. Runge KE, et al. Oocyte CD9 is enriched on the microvillar membrane and required for normal microvillar shape and distribution. Dev Biol. 2007;304:317–325.
    1. Zylkiewicz E, Nowakowska J, Maleszewski M. Decrease in CD9 content and reorganization of microvilli may contribute to the oolemma block to sperm penetration during fertilization of mouse oocyte. Zygote. 2010;18:195–201.
    1. Wolf DP. The block to sperm penetration in zona-free mouse eggs. Developmental Biology. 1978;64:1–10.
    1. Gardner AJ, Williams CJ, Evans JP. Establishment of the mammalian membrane block to polyspermy: evidence for calcium-dependent and -independent regulation. Reproduction. 2007;133:383–393.
    1. Horvath PM, Kellom T, Caulfield J, Boldt J. Mechanistic studies of the plasma membrane block to polyspermy in mouse eggs. Mol Reprod Dev. 1993;34:65–72.
    1. Wortzman-Show GB, Kurokawa M, Fissore RA, Evans JP. Calcium and sperm components in the establishment of the membrane block to polyspermy: studies of ICSI and activation with sperm factor. Mol Hum Reprod. 2007;13:557–565.
    1. Skarnes WC, et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature. 2011;474:337–342.
    1. Nakanishi T, et al. Real-time observation of acrosomal dispersal from mouse sperm using GFP as a marker protein. FEBS Lett. 1999;449:277–283.
    1. Nagy A. Manipulating the Mouse Embryo: A Laboratory Manual. 3rd edn Cold Spring Harbor Laboratory Press; 2003.
    1. Kimura Y, Yanagimachi R. Intracytoplasmic sperm injection in the mouse. Biol Reprod. 1995;52:709–720.
    1. Sun Y, Gallagher-Jones M, Barker C, Wright GJ. A benchmarked protein microarray-based platform for the identification of novel low-affinity extracellular protein interactions. Anal Biochem. 2012;424:45–53.
Methods references
    1. Martin S, et al. Construction of a large extracellular protein interaction network and its resolution by spatiotemporal expression profiling. Mol Cell Proteomics. 2010;9:2654–2665.
    1. Crosnier C, et al. A Library of Functional Recombinant Cell-surface and Secreted P. falciparum Merozoite Proteins. Mol Cell Proteomics. 2013;12:3976–3986.
    1. Durocher Y, Perret S, Kamen A. High-level and high-throughput recombinant protein production by transient transfection of suspension-growing human 293-EBNA1 cells. Nucleic Acids Res. 2002;30:E9.
    1. Golemis EA. Protein-protein Interactions: A Molecular Cloning Manual. Cold Spring Harbor Laboratory Press; 2001.
    1. Kerr JS, Wright GJ. Avidity-based extracellular interaction screening (AVEXIS) for the scalable detection of low-affinity extracellular receptor-ligand interactions. J Vis Exp. 2012:e3881.
    1. Bartholdson SJ, et al. Semaphorin-7A is an erythrocyte receptor for P. falciparum merozoite-specific TRAP homolog, MTRAP. PLoS Pathog. 2012;8:e1003031.

Source: PubMed

3
S'abonner