Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 195
Filter
1.
Eur J Cell Biol ; 102(2): 151296, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36805822

ABSTRACT

The acrosome located within the mammalian sperm head is essential for successful fertilization, as it enables the sperm to penetrate the extracellular layers of the oocyte and fuse with oolemma. However, the mammalian acrosomal vesicle is no longer considered to contain only hydrolytic enzymes. Using label-free nano-scale liquid chromatography tandem mass spectrometry (nLC-MS/MS) proteomics, we identified a total of 885 proteins in the acrosome isolated from spermatozoa obtained from cauda epididymis of free-living house mice Mus musculus musculus contains a total of 885 proteins. Among these, 334 proteins were significantly enriched in the acrosome thus representing 27.3% of the whole proteome of the intact sperm. Importantly, we have detected a total of nine calycins while eight of them belong to the lipocalin protein family. In mice, lipocalins are involved in multi-level chemical communication between individuals including pheromone transport and odor perception. Using an indirect immunofluorescence assay, we demonstrated that lipocalin 5 (LCN5) is expressed in the mouse germ cells, and after completing spermatogenesis, it remains localized in the sperm acrosome until the last step of the extratesticular maturation, the acrosome reaction. The presence of lipocalins in the acrosome and acrosome-reacted sperm suggests their original role as chelators of organic and potentially toxic compounds resulting from ongoing spermiogenesis. Along with this evidence, detected mitochondrial (e.g., a subunit of the cytochrome c oxidase MTCO1) and proteasomal proteins (subunits of both 20 S core proteasome [PSMA2, PSMBs] and 19 S regulatory particle [PSMDs]) in acrosomes provide further evidence that acrosomes could also function as `waste baskets` after testicular sperm maturation.


Subject(s)
Acrosome , Proteomics , Male , Mice , Animals , Acrosome/chemistry , Acrosome/metabolism , Tandem Mass Spectrometry , Semen/metabolism , Spermatozoa/chemistry , Spermatozoa/metabolism , Proteins/metabolism , Lipocalins/analysis , Lipocalins/metabolism , Mammals/metabolism
2.
Reprod Biol ; 20(2): 164-168, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32249155

ABSTRACT

An increasing body of data implicates the Septin family in the pathology of several diseases, including male fertility. The objective of this study was to evaluate the gene and protein expression pattern of Septin 14 in the testis tissue of azoospermic men. In addition, Septin 14 localization was also assessed in the sperm. Testicular tissues were obtained from biopsies of non-obstrutive azoospermic men who underwent diagnostic testicular biopsy in Royan institute and were divided into two groups: TESE + with positive result in testicular sperm extraction (with hypospermatogenesis pathology) and TESE- with negative result (included patients with Sertoli cell only syndrome and maturation arrest pathologies). Total RNA and protein was extracted using trizol reagent. Septin 14 gene and protein expression level were assessed by real-time reverse transcription polymerase chain reaction (RT-PCR) and Western blot techniques, respectively. The localization of Septin 14 protein was also studied by Immunocytochemistry. The expression of Septin 14 was significantly lower (p < 0. 05) in TESE- group than TESE + in both mRNA and protein levels. The localization of Septin 14 protein was detected in the head to tail of normal sperms with high localization in front of the acrosome and the neck. This is a novel localization report on Septin 14 in sperm. Regarding the presence of this protein in the sperm acrosome and neck, it can be concluded that decreasing of Septin 14 protein expression may be associated with the pathogenesis of male infertility and therefore Septin 14 expression level maybe critical for human spermatogenesis.


Subject(s)
Azoospermia/genetics , Gene Expression , Septins/analysis , Septins/genetics , Testis/chemistry , Testis/metabolism , Acrosome/chemistry , Azoospermia/metabolism , Azoospermia/pathology , Biopsy , Humans , Immunohistochemistry , Male , Oligospermia/genetics , Oligospermia/metabolism , Oligospermia/pathology , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction , Spermatogenesis/genetics , Spermatozoa/chemistry , Spermatozoa/ultrastructure , Testis/pathology
3.
Theriogenology ; 149: 46-54, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32234650

ABSTRACT

Sub-fertility is a major problem in crossbred bulls. Identification of subtle differences in the quality of cryopreserved spermatozoa among bulls belonging to different fertility rankings would help determine the latent fertility of semen before their use at field conditions. In the present study, we assessed the status of tyrosine phosphorylation, membrane integrity and acrosome reaction of cryopreserved spermatozoa in crossbred bulls (n = 22) with different levels of field fertility and assessed their relationship with fertility. Bulls were categorized into above-average (n = 4), average (n = 14) and below-average (n = 4) based on their different field fertility rates. The progressive sperm motility was significantly (P < 0.05) higher in above-average fertile bulls compared to either average or below-average fertile bulls whereas sperm membrane integrity and acrosomal reaction status did not differ among the three groups. The proportion of live tyrosine-phosphorylated spermatozoa were significantly (P < 0.05) higher in below-average and average fertile bulls compared to above-average bulls. Immunolocalization of protein tyrosine phosphorylation in spermatozoa revealed that the proportion of spermatozoa showing tyrosine phosphorylation at acrosome and post-acrosomal area (APA) and at acrosome, post-acrosome and tail (APAT) were significantly (P < 0.05) higher in below-average fertile bulls than other groups. The APA pattern (r = -0.605; P < 0.01) and APAT (r = 0.507; P < 0.05) pattern were significantly and negatively correlated with bull fertility. It was concluded that the proportion of live tyrosine-phosphorylated spermatozoa in cryopreserved semen was negatively related to bull fertility.


Subject(s)
Cattle Diseases/physiopathology , Fertility/physiology , Infertility, Male/veterinary , Phosphotyrosine/analysis , Semen Preservation/veterinary , Spermatozoa/physiology , Acrosome/chemistry , Animals , Cattle , Cryopreservation/veterinary , Hybridization, Genetic , Infertility, Male/physiopathology , Male , Sperm Motility/physiology , Sperm Tail/chemistry , Spermatozoa/chemistry , Spermatozoa/ultrastructure
4.
Micron ; 101: 114-122, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28709083

ABSTRACT

The springtail Collembola are characterized by having rolled spermatozoa, with a long cylindrical extracellular structure adhering to the acrosome. This structure is produced by the secretory activity of the testes epithelial cells at almost the end of spermiogenesis. At the beginning of its formation, it is a thin extension with a helical wall and a dense axial region. Later the cylindrical structure shows an inner organization which is different in the several species examined: species of Entomobryidae contain material with a paracrystalline structure, whilst some of Symphypleona contain ovoid structures. The outer envelope of the extracellular structure consists of two overlapped layers orthogonally arranged, clearly identified by cryo-preparations. Immunoblot analysis and lectin stainings have indicated that the cylindrical structure has a glycoproteic composition. As the structure is no longer visible after the sperm transfer into the female spermatheca, it is suggested that it could contain enzymes able to activate the sperm unwinding process and possibly allowing the reacquisition of sperm motility.


Subject(s)
Acrosome/chemistry , Acrosome/ultrastructure , Arthropods/cytology , Glycoproteins/analysis , Multiprotein Complexes/analysis , Spermatozoa/chemistry , Spermatozoa/cytology , Animals , Immunoblotting , Male , Staining and Labeling
5.
Histochem Cell Biol ; 147(1): 27-38, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27539077

ABSTRACT

Peanut agglutinin (PNA), a plant lectin protein that recognizes the galactose ß (1 -> 3) N-acetylgalactosamine carbohydrate sequence, has been widely used as a sperm acrosome-specific marker; however, the acrosomal glycoproteins that specifically bind to PNA have yet to be identified. We herein purified and identified PNA-binding glycoproteins in the mouse testis using biotinylated PNA and streptavidin-coupled magnetic beads, and liquid chromatography-tandem mass spectrometry (LC-MS/MS), respectively. In six repeated experiments, sperm equatorial segment protein 1 (SPESP1) was detected most frequently as a PNA-binding glycoprotein, followed by dipeptidase 3, proacrosin-binding protein, and acrosin prepropeptide. The identification of SPEPS1 in the testis lysate and its PNA-bound fraction was verified with lectin and Western blot analyses, and the co-localization of PNA and SPEPS1 in acrosomes was confirmed with lectin- and immunohistochemistry. Since the PNA reactivity of sperm acrosomes was observed not only in normal mice, but also in SPESP1-deficient mice, although at lower levels, PNA was also considered to bind to other candidate glycoproteins. The present study identified SPESP1 in the acrosome as the primary binding target of PNA in the mouse testis. Further defining the specific lectin-glycoprotein relationships in individual cells will enhance the value of lectin histochemistry.


Subject(s)
Acrosome/metabolism , Carrier Proteins/metabolism , Peanut Agglutinin/metabolism , Seminal Plasma Proteins/metabolism , Testis/metabolism , Acrosome/chemistry , Animals , Carrier Proteins/analysis , Male , Mice , Mice, Inbred C57BL , Peanut Agglutinin/chemistry , Seminal Plasma Proteins/analysis , Testis/chemistry
7.
Adv Anat Embryol Cell Biol ; 220: 1-13, 2016.
Article in English | MEDLINE | ID: mdl-27194347

ABSTRACT

Acrosome reaction is often referred to as acrosomal exocytosis, but it differs significantly from normal exocytosis. While the vesicle membrane initially holding excreting molecules remains on the cell surface during exocytosis, the outer acrosomal membrane and plasma membrane are lost by forming vesicles during acrosome reaction. In this context, the latter process resembles a release of exosome. However, recent experimental data indicate that the most important roles of acrosome reaction lie not in the release of acrosomal contents (or "vesiculated" plasma and outer acrosomal membrane complexes) but rather in changes in sperm membrane. This review describes the mechanism of fertilization vis-a-vis sperm membrane change, with a brief historical overview of the half-century study of acrosome reaction.


Subject(s)
Acrosome Reaction/physiology , Acrosome/physiology , Exocytosis/physiology , Microscopy, Phase-Contrast/history , Sperm Capacitation/physiology , Acrosome/chemistry , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , History, 20th Century , History, 21st Century , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Membrane Fusion , Mice , Mice, Transgenic , Microscopy, Phase-Contrast/instrumentation , Ovum/physiology , Sea Urchins/physiology
8.
Adv Anat Embryol Cell Biol ; 220: 15-33, 2016.
Article in English | MEDLINE | ID: mdl-27194348

ABSTRACT

The acrosome, a single exocytotic vesicle on the head of sperm, has an essential role in fertilization, but the exact mechanisms by which it facilitates sperm-egg interactions remain unresolved. The acrosome contains dozens of secretory proteins that are packaged into the forming structure during spermatogenesis; many of these proteins are localized into specific topographical areas of the acrosome, while others are more diffusely distributed. Acrosomal proteins can also be biochemically classified as components of the acrosomal matrix, a large, relatively insoluble complex, or as soluble proteins. This review focuses on recent findings using genetically modified mice (gene knockouts and transgenic "green acrosome" mice) to study the effects of eliminating acrosomal matrix-associated proteins on sperm structure and function. Some gene knockouts produce infertile phenotypes with obviously missing, specific activities that affect acrosome biogenesis during spermatogenesis or interfere with acrosome function in mature sperm. Mutations that delete some components produce fertile phenotypes with subtler effects that provide useful insights into acrosomal matrix function in fertilization. In general, these studies enable the reassessment of paradigms to explain acrosome formation and function and provide novel, objective insights into the roles of acrosomal matrix proteins in fertilization. The use of genetically engineered mouse models has yielded new mechanistic information that complements recent, important in vivo imaging studies.


Subject(s)
Acrosome/metabolism , Fertilization/physiology , Infertility, Male/genetics , Membrane Proteins/genetics , Peptide Hydrolases/genetics , Acrosome/chemistry , Animals , Female , Gene Expression Regulation , Gene Knockout Techniques , Male , Membrane Fusion , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mutation , Ovum/cytology , Ovum/physiology , Peptide Hydrolases/metabolism , Sperm Maturation/physiology
9.
Adv Anat Embryol Cell Biol ; 220: 71-92, 2016.
Article in English | MEDLINE | ID: mdl-27194350

ABSTRACT

Exocytosis is a fundamental process used by eukaryotic cells to release biological compounds and to insert lipids and proteins in the plasma membrane. Specialized secretory cells undergo regulated exocytosis in response to physiological signals. Sperm exocytosis or acrosome reaction (AR) is essentially a regulated secretion with special characteristics. We will focus here on some of these unique features, covering the topology, kinetics, and molecular mechanisms that prepare, drive, and regulate membrane fusion during the AR. Last, we will compare acrosomal release with exocytosis in other model systems.


Subject(s)
Acrosome Reaction/physiology , Acrosome/metabolism , Cell Membrane/metabolism , Exocytosis/physiology , Acrosome/chemistry , Animals , Calcium/metabolism , Cell Membrane/chemistry , Gene Expression Regulation , Kinetics , Male , Membrane Fusion/physiology , Mice , Phosphatidylinositol Phosphates/metabolism , SNARE Proteins/genetics , SNARE Proteins/metabolism , Signal Transduction , Synaptotagmins/genetics , Synaptotagmins/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab27 GTP-Binding Proteins , rab3 GTP-Binding Proteins/genetics , rab3 GTP-Binding Proteins/metabolism
10.
Adv Anat Embryol Cell Biol ; 220: 107-27, 2016.
Article in English | MEDLINE | ID: mdl-27194352

ABSTRACT

Lipids are critical regulators of mammalian sperm function, first helping prevent premature acrosome exocytosis, then enabling sperm to become competent to fertilize at the right place/time through the process of capacitation, and ultimately triggering acrosome exocytosis. Yet because they do not fit neatly into the "DNA--RNA-protein" synthetic pathway, they are understudied and poorly understood. Here, we focus on three lipids or lipid classes-cholesterol, phospholipids, and the ganglioside G(M1)--in context of the modern paradigm of acrosome exocytosis. We describe how these various- species are precisely segregated into membrane macrodomains and microdomains, simultaneously preventing premature exocytosis while acting as foci for organizing regulatory and effector molecules that will enable exocytosis. Although the mechanisms responsible for these domains are poorly defined, there is substantial evidence for their composition and functions. We present diverse ways that lipids and lipid modifications regulate capacitation and acrosome exocytosis, describing in more detail how removal of cholesterol plays a master regulatory role in enabling exocytosis through at least two complementary pathways. First, cholesterol efflux leads to proteolytic activation of phospholipase B, which cleaves both phospholipid tails. The resultant changes in membrane curvature provide a mechanism for the point fusions now known to occur far before a sperm physically interacts with the zona pellucida. Cholesterol efflux also enables G(M1) to regulate the voltage-dependent cation channel, Ca(V)2.3, triggering focal calcium transients required for acrosome exocytosis in response to subsequent whole-cell calcium rises. We close with a model integrating functions for lipids in regulating acrosome exocytosis.


Subject(s)
Acrosome Reaction/physiology , Acrosome/metabolism , Cholesterol/metabolism , G(M1) Ganglioside/metabolism , Phospholipids/metabolism , Acrosome/chemistry , Acrosome/drug effects , Acrosome Reaction/drug effects , Animals , Calcium/metabolism , Calcium Channels, R-Type/metabolism , Cation Transport Proteins/agonists , Cation Transport Proteins/metabolism , Cholesterol/pharmacology , Enzyme Activation , Exocytosis/drug effects , Female , G(M1) Ganglioside/pharmacology , Lysophospholipase/metabolism , Male , Membrane Fusion/drug effects , Membrane Fusion/physiology , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Phospholipids/pharmacology , Sperm Capacitation/drug effects , Sperm Capacitation/physiology , Zona Pellucida/physiology
11.
Adv Anat Embryol Cell Biol ; 220: 129-44, 2016.
Article in English | MEDLINE | ID: mdl-27194353

ABSTRACT

Mammalian sperm require to undergo an exocytotic process called acrosomal exocytosis in order to be able to fuse with the oocyte. This ability is acquired during the course of sperm capacitation. This review is focused on one aspect related to this acquisition: the role of the actin cytoskeleton. Evidence from different laboratories indicates that actin polymerization occurs during capacitation, and the detection of several actin-related proteins suggests that the cytoskeleton is involved in important sperm functions. In other mammalian cells, the cortical actin network acts as a dominant negative clamp which blocks constitutive exocytosis but, at the same time, is necessary to prepare the cell to undergo regulated exocytosis. Thus, F-actin stabilizes structures generated by exocytosis and supports the physiological progression of this process. Is this also the case in mammalian sperm? This review summarizes what is currently known about actin and its related proteins in the male gamete, with particular emphasis on their role in acrosomal exocytosis.


Subject(s)
Acrosome Reaction/genetics , Acrosome/metabolism , Actin Cytoskeleton/genetics , Actins/genetics , Exocytosis/genetics , Sperm Capacitation/genetics , Acrosome/chemistry , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Actins/chemistry , Actins/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Humans , Lim Kinases/genetics , Lim Kinases/metabolism , Male , Mice , Phospholipase D/genetics , Phospholipase D/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Signal Transduction
12.
Adv Anat Embryol Cell Biol ; 220: 159-72, 2016.
Article in English | MEDLINE | ID: mdl-27194355

ABSTRACT

The acrosome reaction (AR) is a universal requisite for sperm-egg fusion. However, whereas through the animal kingdom fusion of spermatozoa with the egg plasma membrane occurs via the inner acrosomal membrane exposed after the AR, in eutherian mammals, gamete fusion takes place through a specialized region of the acrosome known as the equatorial segment (ES) which becomes fusogenic only after the AR is completed. This chapter focuses on the different molecular mechanisms involved in the acquisition of the fusogenicity of the ES after the AR. We provide an update of the knowledge about the proteins proposed to have a role in this process either by modifying cytoskeletal and/or membrane molecules or by relocalizing to the ES after the AR to subsequently participate in gamete fusion.


Subject(s)
Acrosome Reaction/genetics , Acrosome/metabolism , Membrane Fusion/genetics , Sperm Capacitation/genetics , Zona Pellucida/physiology , Acrosin/genetics , Acrosin/metabolism , Acrosome/chemistry , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Female , Gene Expression Regulation , Immunoglobulins/genetics , Immunoglobulins/metabolism , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Seminal Plasma Proteins/genetics , Seminal Plasma Proteins/metabolism , Signal Transduction
13.
Cell Tissue Res ; 363(1): 147-157, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26271197

ABSTRACT

The acrosome is not just a bag of enzymes, most of which, if not all, are singly non-essential for sperm-oocyte interaction. The Golgi-derived acrosomal cap reveals some extraordinary development and structure particularities. The acrosome of eutherian spermatozoa basically consists of two parts, the anterior and equatorial segments; the present review is devoted to the former, the initial actor in fertilization. Its occasional fanciful morphological changes during epididymal maturation are analyzed, together with its heterogeneous contents: enzymes, zona binding proteins, structural proteins (matrix) and yet to be chemically characterized crystalloids. The plasma and acrosomal membranes present stabilized ordered domains, whereas glycoprotein-free areas appear during capacitation and before fusion. Exocytosis, induced by the cumulus oophorus and/or the zona pellucida, may generally start proximally and progress anteriorly, resulting in the detachment of a hybrid membrane shroud, whose entity is probably maintained by the bound matrix. Immediately released soluble enzymes must be active during the first interactions of the gametes, whereas other lysins, bound to the matrix or stored as proenzymes, are only progressively released. Zona binding is probably achieved via the shroud and/or the IAM (depending on species). Penetration along an incurved slit through the stratified zona is allowed by the rigid and denuded head tip and flagellar hyperactivity, and assisted by the local proteolytic activity of proteasomes bound to the IAM, the unique essential zona lysin system.


Subject(s)
Acrosome/metabolism , Mammals/metabolism , Acrosome/chemistry , Acrosome/ultrastructure , Acrosome Reaction , Animals , Crystallization , Fertilization , Humans , Male , Mammals/classification , Sperm Maturation , Sperm-Ovum Interactions , Zona Pellucida/metabolism
14.
Sci Rep ; 5: 15321, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26470849

ABSTRACT

All cells are covered by glycans, an individually unique layer of oligo- and polysaccharides that are critical moderators of self-recognition and other cellular-level interactions (e.g. fertilization). The functional similarity between these processes suggests that gamete surface glycans may also have an important, but currently overlooked, role in sexual selection. Here we develop a user-friendly methodological approach designed to facilitate future tests of this possibility. Our proposed method is based on flow cytometric quantification of female-induced sperm acrosome reaction and sperm surface glycan modifications in the Mediterranean mussel Mytilus galloprovincialis. In this species, as with many other taxa, eggs release water-soluble factors that attract conspecific sperm (chemoattraction) and promote potentially measurable changes in sperm behavior and physiology. We demonstrate that flow cytometry is able to identify sperm from other seawater particles as well as accurately measure both acrosome reaction and structural modifications in sperm glycans. This methodological approach can increase our understanding of chemically-moderated gamete-level interactions and individual-specific gamete recognition in Mytilus sp. and other taxa with similar, easily identifiable acrosome structure. Our approach is also likely to be applicable to several other species, since carbohydrate-mediated cellular-level interactions between gametes are universal among externally and internally fertilizing species.


Subject(s)
Acrosome/metabolism , Lectins/metabolism , Polysaccharides/metabolism , Spermatozoa/physiology , Acrosome/chemistry , Acrosome Reaction , Animals , Bivalvia , Female , Fertilization , Flow Cytometry , Lectins/chemistry , Male , Monosaccharides/chemistry , Monosaccharides/metabolism , Polysaccharides/chemistry
15.
Acta Biochim Biophys Sin (Shanghai) ; 47(11): 861-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26363282

ABSTRACT

ß-defensins, preferentially expressed in male reproductive tracts, particularly in the testes and epididymis with region-specific patterns, play an important role in both innate immunity and sperm fertility. Expressed in the caput region of epididymis, ß-defensins have been known to contribute to innate immunity, sperm motility initiation, and maintenance. However, ß-defensins of the initial region remain to be uncharacterized. In this study, rat ß-defensin 42 (Defb42) was revealed to be exclusively located in the principal cells at the initial segment of the rat epididymis and its sperm's acrosome. Furthermore, the expression of Defb42 was dependent on luminal testicular factors and developmental phases. The recombinant Defb42 was predominantly antimicrobial not against Candida albicans, but against Escherichia coli and Staphylococcus aureus. Based on these findings, Defb42 was suggested to play a dual role in sperm fertility and host defense in rat epididymis.


Subject(s)
Epididymis/metabolism , Sperm Motility , beta-Defensins/immunology , Acrosome/chemistry , Animals , Candida albicans/physiology , Epididymis/anatomy & histology , Epididymis/immunology , Escherichia coli/physiology , Male , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Staphylococcus aureus/physiology , Testosterone/administration & dosage , beta-Defensins/analysis , beta-Defensins/genetics
16.
J Reprod Dev ; 61(6): 519-24, 2015.
Article in English | MEDLINE | ID: mdl-26300347

ABSTRACT

The purposes of this study were to examine the relationship between male artificial insemination (AI) fertility and sperm acrosomal conditions assessed by new and conventional staining techniques and to identify possible reproductive dysfunctions causing low conception rates in AI using frozen-thawed spermatozoa with poor acrosomal conditions in Japanese Black bulls. We investigated individual differences among bulls in the results concerning (1) acrosomal conditions of frozen-thawed spermatozoa as assessed by not merely peanut agglutinin-lectin staining (a conventional staining technique) but also immunostaining of acrosomal tyrosine-phosphorylated proteins (a new staining technique), (2) routine AI using frozen-thawed spermatozoa as assessed by pregnancy diagnosis, (3) in vivo fertilization of frozen-thawed spermatozoa and early development of fertilized eggs as assessed by superovulation/AI-embryo collection tests and (4) in vitro fertilization of frozen-thawed spermatozoa with oocytes. The percentages of frozen-thawed spermatozoa with normal acrosomal conditions assessed by the abovementioned staining techniques were significantly correlated with the conception rates of routine AI, rates of transferable embryos in superovulation/AI-embryo collection tests and in vitro fertilization rates. These results are consistent with new suggestions that the distribution of acrosomal tyrosine-phosphorylated proteins as well as the acrosomal morphology of frozen-thawed spermatozoa are AI fertility-associated markers that are valid for the prediction of AI results and that low conception rates in AI using frozen-thawed spermatozoa with poor acrosomal conditions result from reproductive dysfunctions in the processes between sperm insemination into females and early embryo development, probably failed fertilization of frozen-thawed spermatozoa with oocytes.


Subject(s)
Acrosome/physiology , Cattle/physiology , Insemination, Artificial/veterinary , Spermatozoa/physiology , Acrosome/chemistry , Animals , Female , Fertility/physiology , Freezing , Infertility/veterinary , Insemination, Artificial/methods , Lectins , Male , Organophosphates , Peanut Agglutinin , Polymers , Proteins/analysis
17.
J Reprod Dev ; 61(3): 205-10, 2015.
Article in English | MEDLINE | ID: mdl-25754239

ABSTRACT

Cryopreservation of boar semen is still considered suboptimal due to lower fertility as compared with fresh samples when glycerol, a permeating cryoprotectant, is used. Trehalose is a non-permeable cryoprotectant and nonreducing disaccharide known to stabilize proteins and biologic membranes. The aim of this study was to evaluate the cryosurvival and in vitro penetrability of boar spermatozoa when glycerol was replaced with trehalose in a freezing extender. Ejaculated Berkshire semen samples were diluted in egg yolk-based freezing extender containing glycerol (100 mM) or trehalose (0, 50, 100, 150, 200 and 250 mM) and cryopreserved using a straw freezing procedure. Thawed samples were analyzed for motility, viability, mitochondrial membrane potential (MMP), and acrosome integrity. In experiment 2, penetrability of spermatozoa cryopreserved with 100 mM glycerol or trehalose was examined. Replacement of cryoprotectant glycerol (100 mM) with trehalose had no effect on sperm viability, but replacing it with 100 mM trehalose improved motility, MMP and acrosome integrity significantly. Sperm motility and MMP were considerably higher in 100 mM trehalose, whereas the acrosome integrity was substantially higher in 100-250 mM trehalose. The in vitro penetration rate was also significantly higher in spermatozoa cryopreserved with trehalose (61.3%) than in those cryopreserved with glycerol (43.6%). In conclusion, 100 mM non-permeable trehalose can be used to replace glycerol, a permeating cryoprotectant, for maintenance of better post-thaw quality of boar spermatozoa.


Subject(s)
Cryopreservation , Glycerol/chemistry , Semen Preservation/methods , Spermatozoa/physiology , Trehalose/chemistry , Acrosome/chemistry , Animals , Cell Survival , Cryoprotective Agents/chemistry , Fertilization in Vitro , Male , Membrane Potential, Mitochondrial , Oocytes/cytology , Semen , Sperm Motility , Swine
18.
Mol Reprod Dev ; 81(4): 315-25, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24415168

ABSTRACT

Oocyte meiotic maturation is a developmental transition that starts during germinal-vesicle breakdown and ends at the arrest in metaphase of meiosis II. This transition is associated with changes to both the proteins that are synthesized and the abundance/distribution of post-translational modifications that are crucial for subsequent fertilization and embryogenesis. Here, we isolated and cultured rat oocytes in vitro during both metaphase of meiosis I (MI) and meiosis II (MII) stages, respectively, and then compared their proteomic profiles by high-resolution, two-dimensional gel electrophoresis (2DE) followed by mass spectrometry. We found that the expression of five proteins was up-regulated while six proteins were down-regulated when comparing MI to MII oocytes. The expression of ERp57, an endoplasmic reticulum chaperone, underwent a dramatic increase between MI and MII oocytes, and became concentrated in a dome-shaped area of the cell surface within the microvillar region. A similar profile was observed during spermatogenesis, and sperm ERp57 eventually localized to the head and flagellum surfaces, finally ending in the equatorial region of acrosome-reacted sperm. Given the localization pattern, we tested and found that a polyclonal antiserum created against recombinant rat ERp57 significantly inhibited spermatozoa from penetrating zona pellucida-free oocytes without affecting either sperm motility or the acrosome reaction. These results indicate that ERp57 expression on oocytes, and possibly sperm, plays an important physiological role during sperm-egg fusion.


Subject(s)
Meiosis , Oocytes/metabolism , Protein Disulfide-Isomerases/physiology , Sperm-Ovum Interactions/physiology , Acrosome/chemistry , Animals , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Female , Fertilization in Vitro , Gene Expression Profiling , Immune Sera , Male , Mass Spectrometry , Membrane Fusion , Microvilli/chemistry , Oocytes/growth & development , Protein Disulfide-Isomerases/antagonists & inhibitors , Protein Disulfide-Isomerases/biosynthesis , Protein Disulfide-Isomerases/genetics , Proteomics , Rats , Recombinant Proteins/immunology , Subcellular Fractions/chemistry
19.
Reproduction ; 147(2): 231-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24277869

ABSTRACT

It has been recently shown in mice that sperm undergo acrosome reaction (AR) by passing through cumulus cells; furthermore, the acrosome-reacted sperm can bind to zona pellucida and consequently fertilise the egg. During AR, the relocation of the primary fusion protein IZUMO1 into the equatorial segment is crucial for sperm-egg fusion. There is a high rate of spontaneous AR in rodents, with up to 60% in promiscuous species. The aim of this study was to clarify whether the IZUMO1 relocation in sperm after spontaneous and induced AR is the same, and whether there is a correlation between the speed of IZUMO1 relocation and species-specific mating behaviour in field mice. Immunofluorescent detection of IZUMO1 dynamics during the in vitro capacitation, spontaneous, calcium ionophore and progesterone-induced AR was monitored. Our results show that during spontaneous AR, there is a clear IZUMO1 relocation from the acrosomal cap to the equatorial segment, and further over the whole sperm head. In addition, there is positive tail tyrosine phosphorylation (TyrP) associated with hyperactive motility. Moreover, the beginning and the progress of IZUMO1 relocation and tail TyrP positively correlate with the level of promiscuity and the acrosome instability in promiscuous species. The findings that crucial molecular changes essential for sperm-egg fusion represented by dynamic movements of IZUMO1 also happen during spontaneous AR are vital for understanding fertilisation in mice.


Subject(s)
Acrosome Reaction/physiology , Immunoglobulins/metabolism , Membrane Proteins/metabolism , Murinae/physiology , Spermatozoa/chemistry , Spermatozoa/physiology , Acrosome/chemistry , Acrosome Reaction/drug effects , Animals , Immunoglobulins/analysis , Male , Membrane Proteins/analysis , Mice , Mice, Inbred BALB C , Progesterone/pharmacology , Sexual Behavior, Animal , Species Specificity , Sperm Capacitation , Sperm-Ovum Interactions/physiology , Zona Pellucida/metabolism
20.
Mol Reprod Dev ; 80(6): 488-502, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23661501

ABSTRACT

Sperm CRISP2 has been proposed to be involved in sperm-egg fusion. After the acrosome reaction, it appears at the equatorial segment (EqS) of human sperm; the mechanism underlying the appearance of CRISP2 at the EqS remains unknown, though. Here, we provide evidence showing the re-association of sperm acrosomal CRISP2 at the EqS during the acrosome reaction. Results showed that F-actin is not involved in the relocalization of CRISP2. We found that basic, but not acidic, conditions can solubilize CRISP2 from sperm cells, suggesting that CRISP2 is a component of the acrosome and that it is released from the acrosome during the acrosome reaction. Purified, biotinylated human sperm acrosomal CRISP2 binds to the EqS of acrosome-reacted sperm in a dose-dependent manner, revealing that CRISP2 detected at the EqS of acrosome-reacted sperm comes from the population stored in the acrosome. The association of CRISP2 at the EqS is very strong, and does not depend on ionic interactions or intermolecular disulfide bonds. Interestingly, the restriction of CRISP2 at the EqS was diminished when EGTA was present in the media, indicating that Ca(2+) is required for maintaining CRISP2 at the EqS. This study supports the possibility that CRISP2 may help modify the EqS membrane to make this domain fusion-competent.


Subject(s)
Acrosome Reaction/physiology , Acrosome/metabolism , Glycoproteins/metabolism , Acrosome/chemistry , Acrosome/drug effects , Acrosome Reaction/drug effects , Actins/metabolism , Adult , Calcimycin/pharmacology , Cell Adhesion Molecules , Glycoproteins/analysis , Glycoproteins/chemistry , Humans , Ionophores/pharmacology , Male , Microscopy, Fluorescence , Protein Stability , Spermatozoa/chemistry , Spermatozoa/drug effects , Spermatozoa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...