Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Anim Sci Technol ; 63(1): 58-68, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33987584

ABSTRACT

Several herbs including Artemisia are known to possess conceptive property. In the present study, mouse spermatozoa were incubated with ethanol extract of Artemisia vulgaris leaves. The effect of extract on acrosome exocytosis was studied by labeling spermatozoa with fluorescein isothiocyanate (FITC) peanut agglutinin and by staining with Coomassie blue. Viability and membrane integrity were studied by Trypan-blue staining and hypo-osmotic swelling test. Artemisia extract at very low concentration caused precocious acrosome reaction and loss of sperm viability. Acrosome reaction increased remarkably from 22.63% to 88.42% with increasing extract concentration from 0 to 2,000 µg/mL. However, the viability loss of spermatozoa was increased from 11.71% in control to 63.73% in samples treated, evaluated by Trypan-blue staining method. Membrane damage caused by the extract, evaluated by hypo-osmotic swelling test was even low, ranging from 2.27% to only 24.23%. These results indicate that Artemisia extract might block fertilization by causing precocious acrosome exocytosis in spermatozoa. A direct contraceptive effect was tested by injecting the plant extract into the vagina of female mice and then allowing them to mate with normal males. The treated female mice delivered significantly fewer litters in comparison to the control.

2.
Cell Tissue Res ; 379(3): 561-576, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31897834

ABSTRACT

Defective mammalian spermatozoa are marked on their surface by proteolytic chaperone ubiquitin. To identify potential ubiquitinated substrates in the defective spermatozoa, we resolved bull sperm protein extracts on a two-dimensional gel and isolated a 64-65-kDa spot (p64) corresponding to one of the major ubiquitin-immunoreactive bands observed in the one-dimensional Western blots. Immune serum raised against this protein recognized a prominent, possibly glycosylated band/spot in the range of 55-68 kDa, consistent with the original spot used for immunization. Internal sequences obtained by Edman degradation of this spot matched the sequence of arylsulfatase A (ARSA), the sperm acrosomal enzyme thought to be important for fertility. By immunofluorescence, a prominent signal was detected on the acrosomal surface (boar and bull) and on the sperm tail principal piece (bull). A second immune serum raised against a synthetic peptide corresponding to an immunogenic internal sequence (GTGKSPRRTL) of the porcine ARSA also labeled sperm acrosome and principal piece. Both sera showed diminished immunoreactivity in the defective bull spermatozoa co-labeled with an anti-ubiquitin antibody. Western blotting and image-based flow cytometry (IBFC) confirmed a reduced ARSA immunoreactivity in the immotile sperm fraction rich in ubiquitinated spermatozoa. Larger than expected ARSA-immunoreactive bands were found in sperm protein extracts immunoprecipitated with anti-ubiquitin antibodies and affinity purified with matrix-bound, recombinant ubiquitin-binding UBA domain. These bands did not show the typical pattern of ARSA glycosylation but overlapped with bands preferentially binding the Lens culinaris agglutinin (LCA) lectin. By both epifluorescence microscopy and IBFC, the LCA binding was increased in the ubiquitinated spermatozoa with diminished ARSA immunoreactivity. ARSA was also found in the epididymal fluid suggesting that in addition to intrinsic ARSA expression in the testis, epididymal spermatozoa take up ARSA on their surface during the epididymal passage. We conclude that sperm surface ARSA is one of the ubiquitinated sperm surface glycoproteins in defective bull spermatozoa. Defective sperm surface thus differs from normal sperm surface by increased ubiquitination, reduced ARSA binding, and altered glycosylation.


Subject(s)
Cerebroside-Sulfatase/metabolism , Spermatozoa/metabolism , Ubiquitin/biosynthesis , Animals , Cattle , Immunohistochemistry , Male , Microscopy, Fluorescence , Spermatozoa/enzymology , Spermatozoa/pathology , Sus scrofa , Ubiquitin/metabolism
3.
PLoS One ; 8(5): e61000, 2013.
Article in English | MEDLINE | ID: mdl-23734172

ABSTRACT

Spermatid specific thioredoxin-3 (SPTRX3 or TXNDC8) is a testis/male germ line specific member of thioredoxin family that accumulates in the superfluous cytoplasm of defective human spermatozoa. We hypothesized that semen levels of SPTRX3 are reflective of treatment outcome in assisted reproductive therapy (ART) couples treated by in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI). Relationship between SPTRX3 and treatment outcome was investigated in 239 couples undergoing ART at an infertility clinic. Sperm content of SPTRX3 was evaluated by flow cytometry and epifluorescence microscopy, and correlated with clinical semen analysis parameters, and data on embryo development and pregnancy establishment. High SPTRX3 levels (>15% SPTRX3-positive spermatozoa) were found in 51% of male infertility patients (n = 72), in 20% of men from couples with unexplained, idiopathic infertility (n = 61) and in 14% of men from couples previously diagnosed with female-only infertility (n = 85). Couples with high SPTRX3 produced fewer two-pronuclear zygotes and had a reduced pregnancy rate (19.2% pregnant with >15% SPTRX3-positive spermatozoa vs. 41.2% pregnant with <5% SPTRX3-positive sperm; one-sided p<0.05). The average pregnancy rate of all 239 couples was 25.1%. Live birth rate was 19.2% and lowest average SPTRX3 levels were found in couples that delivered twins. Men with >15% of SPTRX3-positive spermatozoa, a cutoff value established by ROC analysis, had their chance of fathering children by IVF or ICSI reduced by nearly two-thirds. The percentage of SPTRX3-positive spermatozoa had predictive value for pregnancy after ART. Gradient purification and sperm swim-up failed to remove all SPTRX3-positive spermatozoa from semen prepared for ART. In summary, the elevated semen content of SPTRX3 in men from ART couples coincided with reduced incidence of pregnancy by IVF or ICSI, identifying SPTRX3 as a candidate biomarker reflective of ART outcome.


Subject(s)
Infertility, Male/therapy , Reproductive Techniques, Assisted , Semen/metabolism , Spermatids/metabolism , Thioredoxins/metabolism , Adult , Biomarkers/metabolism , Blotting, Western , Female , Fertilization in Vitro , Flow Cytometry , Humans , Infant, Newborn , Infertility, Male/metabolism , Male , Microscopy, Fluorescence , Pregnancy , Pregnancy Outcome , Pregnancy Rate , Regression Analysis , Semen/cytology , Semen Analysis , Sperm Injections, Intracytoplasmic
4.
PLoS One ; 6(2): e17256, 2011 Feb 23.
Article in English | MEDLINE | ID: mdl-21383844

ABSTRACT

Despite decades of research, the mechanism by which the fertilizing spermatozoon penetrates the mammalian vitelline membrane, the zona pellucida (ZP) remains one of the unexplained fundamental events of human/mammalian development. Evidence has been accumulating in support of the 26S proteasome as a candidate for echinoderm, ascidian and mammalian egg coat lysin. Monitoring ZP protein degradation by sperm during fertilization is nearly impossible because those few spermatozoa that penetrate the ZP leave behind a virtually untraceable residue of degraded proteins. We have overcome this hurdle by designing an experimentally consistent in vitro system in which live boar spermatozoa are co-incubated with ZP-proteins (ZPP) solubilized from porcine oocytes. Using this assay, mimicking sperm-egg interactions, we demonstrate that the sperm-borne proteasomes can degrade the sperm receptor protein ZPC. Upon coincubation with motile spermatozoa, the solubilized ZPP, which appear to be ubiquitinated, adhered to sperm acrosomal caps and induced acrosomal exocytosis/formation of the acrosomal shroud. The degradation of the sperm receptor protein ZPC was assessed by Western blotting band-densitometry and proteomics. A nearly identical pattern of sperm receptor degradation, evident already within the first 5 min of coincubation, was observed when the spermatozoa were replaced with the isolated, enzymatically active, sperm-derived proteasomes. ZPC degradation was blocked by proteasomal inhibitors and accelerated by ubiquitin-aldehyde(UBAL), a modified ubiquitin protein that stimulates proteasomal proteolysis. Such a degradation pattern of ZPC is consistent with in vitro fertilization studies, in which proteasomal inhibitors completely blocked fertilization, and UBAL increased fertilization and polyspermy rates. Preincubation of intact zona-enclosed ova with isolated active sperm proteasomes caused digestion, abrasions and loosening of the exposed zonae, and significantly reduced the fertilization/polyspermy rates after IVF, accompanied by en-mass detachment of zona bound sperm. Thus, the sperm borne 26S proteasome is a candidate zona lysin in mammals. This new paradigm has implications for contraception and assisted reproductive technologies in humans, as well as animals.


Subject(s)
Fertilization/physiology , Proteasome Endopeptidase Complex/metabolism , Receptors, Cell Surface/metabolism , Spermatozoa/metabolism , Zona Pellucida/metabolism , Amino Acid Sequence , Animals , Egg Proteins/chemistry , Egg Proteins/metabolism , Female , Male , Mammals/metabolism , Mammals/physiology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Models, Biological , Molecular Sequence Data , Proteasome Endopeptidase Complex/physiology , Protein Processing, Post-Translational/physiology , Receptors, Cell Surface/chemistry , Sperm-Ovum Interactions/physiology , Swine , Zona Pellucida Glycoproteins
5.
Proteomics Clin Appl ; 4(3): 337-51, 2010 Mar.
Article in English | MEDLINE | ID: mdl-21137054

ABSTRACT

PURPOSE: Identification of the biomarkers of oocyte quality, and developmental and reprogramming potential is of importance to assisted reproductive technology in humans and animals. EXPERIMENTAL DESIGN: PerkinElmer ExacTag™ Kit was used to label differentially proteins in pig oocyte extracts (oocyte proteome) and pig oocyte-conditioned in vitro maturation media (oocyte secretome) obtained with high- and low-quality oocytes. RESULTS: We identified 16 major proteins in the oocyte proteome that were expressed differentially in high- versus low-quality oocytes. More abundant proteins in the high-quality oocyte proteome included kelch-like ECH-associated protein 1 (an adaptor for ubiquitin-ligase CUL3), nuclear export factor CRM1 and ataxia-telangiectasia mutated protein kinase. Dystrophin (DMD) was more abundant in low-quality oocytes. In the secretome, we identified 110 proteins, including DMD and cystic fibrosis transmembrane conductance regulator, two proteins implicated in muscular dystrophy and cystic fibrosis, respectively. Monoubiquitin was identified in the low-quality-oocyte secretome. CONCLUSIONS AND CLINICAL IMPLICATIONS: A direct, quantitative proteomic analysis of small oocyte protein samples can identify potential markers of oocyte quality without the need for a large amount of total protein. This approach will be applied to discovery of non-invasive biomarkers of oocyte quality in assisted human reproduction and in large animal embryo transfer programs.


Subject(s)
Oocytes/metabolism , Proteomics/methods , Animals , Biomarkers/metabolism , Female , Gene Expression Regulation , Humans , Proteome/metabolism , Swine
6.
Cell Tissue Res ; 341(2): 325-40, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20526895

ABSTRACT

Proteolysis of ubiquitinated sperm and oocyte proteins by the 26S proteasome is necessary for the success of mammalian fertilization, including but not limited to acrosomal exocytosis and sperm-zona pellucida (ZP) penetration. The present study examined the role of PSMD4, an essential non-ATPase subunit of the proteasomal 19S regulatory complex responsible for proteasome-substrate recognition, in sperm-ZP penetration during porcine fertilization in vitro (IVF). Porcine sperm-ZP penetration, but not sperm-ZP binding, was blocked in the presence of a monoclonal anti-PSMD4 antibody during IVF. Inclusion in the fertilization medium of mutant ubiquitins (Ub+1 and Ub5+1), which are refractory to processing by the 19S regulatory complex and associated with Alzheimer's disease, also inhibited fertilization. This observation suggested that subunit PSMD4 is exposed on the sperm acrosomal surface, a notion that was further supported by the binding of non-cell permeant, biotinylated proteasomal inhibitor ZL3VS to the sperm acrosome. Immunofluorescence localized PSMD4 in the sperm acrosome. Immunoprecipitation and proteomic analysis revealed that PSMD4 co-precipitated with porcine sperm-associated acrosin inhibitor (AI). Ubiquitinated species of AI were isolated from boar sperm extracts by affinity purification of ubiquitinated proteins using the recombinant UBA domain of p62 protein. Some proteasomes appeared to be anchored to the sperm head inner acrosomal membrane, as documented by co-fractionation studies. In conclusion, the 19S regulatory complex subunit PSMD4 is involved in the sperm-ZP penetration during fertilization. The recognition of substrates on the ZP by the 19S proteasomal regulatory complex is essential for the success of porcine/mammalian fertilization in vitro.


Subject(s)
Proteasome Inhibitors , Sperm-Ovum Interactions , Spermatozoa/enzymology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Blotting, Western , Carrier Proteins/immunology , Fertilization in Vitro/drug effects , Male , Molecular Sequence Data , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism , Proteomics , Seminal Vesicle Secretory Proteins/immunology , Sperm-Ovum Interactions/drug effects , Swine , Trypsin Inhibitor, Kazal Pancreatic/immunology , Ubiquitinated Proteins/metabolism
7.
Cell Tissue Res ; 340(3): 569-81, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20449608

ABSTRACT

Mammalian spermatozoa complete their morphogenesis and acquire their fertilizing potential in the epididymis. Prominent among the hallmarks of epididymal sperm maturation is the proximal-distal migration of the cytoplasmic droplet (CD), the last remnant of the spermatogenic cell cytoplasm, down the sperm flagellum. Failure to shed the CD has been associated with male infertility. Because of the presence of the organelle degradation enzyme 15-lipoxygenase (15LOX) in sperm CD, we hypothesize that subfertile male Alox15 mice lacking the 15Lox gene display sperm CD anomalies. Caput and cauda epididymal sperm samples from seven adult Alox15 and seven wild-type (wt) males of equal age were examined by differential interference contrast microscopy (DIC) and transmission electron microscopy (TEM). Compared with wt males, Alox15 males had significantly more spermatozoa with a retained CD in both caput (P = 0.004) and cauda (P = 0.005) epididymidis. TEM and DIC analyses revealed intact mitochondria present in the CDs of epididymal Alox15 spermatozoa. The CDs of wt spermatozoa, however, had a smooth appearance and contained only hollow membrane vesicles, with no intact mitochondria embedded in their CD matrix. Epithelial lesions, phagocytosis-like figures, and missing or aberrant apical blebs were observed in the caput epididymidis of Alox15 males. Thus, the process of epididymal sperm maturation and CD migration is altered in Alox15 males. Aberrant sperm maturation might contribute to the reduced fertility and smaller litter size of Alox15 mice, a rare example of subfertile mutants displaying normal spermatogenesis but altered epididymal sperm maturation.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Cytoplasm/metabolism , Epididymis/pathology , Infertility, Male/enzymology , Infertility, Male/physiopathology , Multienzyme Complexes/metabolism , Sperm Maturation/physiology , Sperm Motility/physiology , Animals , Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 15-Lipoxygenase/deficiency , Epididymis/enzymology , Epididymis/ultrastructure , Epithelium/enzymology , Epithelium/pathology , Epithelium/ultrastructure , Male , Mice , Mice, Inbred C57BL , Multienzyme Complexes/deficiency , Spermatogenesis , Spermatozoa/enzymology , Spermatozoa/pathology , Spermatozoa/ultrastructure
8.
J Reprod Immunol ; 84(2): 154-63, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20004025

ABSTRACT

The 26S proteoasome is a multi-subunit protease specific to ubiquitinated substrate proteins. It is composed of a 20S proteasomal core with substrate degradation activity, and a 19S regulatory complex that acts in substrate recognition, deubiquitination, priming and transport to the 20S core. Inhibition of proteolytic activities associated with the sperm acrosome-borne 20S core prevents fertilization in mammals, ascidians and echinoderms. Less is known about the function of the proteasomal 19S complex during fertilization. The present study examined the role of PSMD8, an essential non-ATPase subunit of the 19S complex, in sperm-ZP penetration during porcine fertilization in vitro (IVF). Immunofluorescence localized PSMD8 to the outer acrosomal membrane, acrosomal matrix and the inner acrosomal membrane. Colloidal gold transmission electron microscopy detected PSMD8 on the surface of vesicles in the acrosomal shroud, formed as a result of zona pellucida-induced acrosomal exocytosis. Contrary to the inhibition of fertilization by blocking of the 20S core activities, fertilization and polyspermy rates were increased by adding anti-PSMD8 antibody to fertilization medium. This observation is consistent with a possible role of PSMD8 in substrate deubiquitination, a process which when blocked, may actually accelerate substrate proteolysis by the 26S proteasome. Subunit PSMD8 co-immunoprecipitated with acrosomal surface-associated spermadhesin AQN1. This association indicates that the sperm acrosome-borne proteasomes become exposed onto the sperm surface following the acrosomal exocytosis. Since immunological blocking of subunit PSMD8 increases the rate of polyspermy during porcine fertilization, the activity of the 19S complex may be a rate-limiting factor contributing to anti-polyspermy defense during porcine fertilization.


Subject(s)
Acrosome/metabolism , Oocytes/metabolism , Proteasome Endopeptidase Complex/metabolism , Zona Pellucida/metabolism , Acrosome/immunology , Acrosome/ultrastructure , Animals , Antibodies/pharmacology , Cells, Cultured , Exocytosis , Female , Fertilization , Fertilization in Vitro/drug effects , Male , Microscopy, Electron, Transmission , Oocytes/cytology , Proteasome Endopeptidase Complex/immunology , Protein Binding , Seminal Plasma Proteins/metabolism , Sperm-Ovum Interactions/drug effects , Swine , Ubiquitinated Proteins/metabolism , Zona Pellucida/immunology
9.
Biol Reprod ; 80(6): 1168-77, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19208552

ABSTRACT

Peroxiredoxin 2 (PRDX2) is a highly efficient redox protein that neutralizes hydrogen peroxide, resulting in protection of cells from oxidative damage and in regulation of peroxide-mediated signal transduction events. The oxidized form of PRDX2 is reverted back to the reduced form by the thioredoxin system. In the present study, we investigated the presence of PRDX2 in mouse and boar spermatozoa and in mouse spermatids using proteomic techniques and immunocytochemistry. Sperm and spermatid extracts displayed a 20-kDa PRDX2 band on Western blotting. PRDX2 occurred as a Triton-soluble form in spermatids and as a Triton-insoluble form in mature spermatozoa. Boar seminiferous tubule extracts were immunoprecipitated with PRDX2 antibody and separated by SDS-PAGE. Peptide mass fingerprinting by matrix-assisted laser desorption ionization-time of flight (TOF) and microsequencing by nanospray quadrupole-quadrupole TOF tandem mass spectrometry revealed the presence of PRDX2 ions in the immunoprecipitated band, along with sperm mitochondria-associated cysteine-rich protein, cellular nucleic acid-binding protein, and glutathione peroxidase 4. In mouse spermatocytes and spermatids, diffuse labeling of PRDX2 was observed in the cytoplasm and residual bodies. After spermiation, PRDX2 localization became confined to the mitochondrial sheath of the sperm tail midpiece. Boar spermatozoa displayed similar PRDX2 localization as in mouse spermatozoa. Boar spermatozoa with disrupted acrosomes expressed PRDX2 in the postacrosomal sheath region. Peroxidase enzyme activity of boar sperm extracts was evaluated by estimating the rate of NADPH oxidation in the presence or absence of a glutathione depletor (diethyl maleate) or a glutathione reductase inhibitor (carmustine). Diethyl maleate partially inhibited peroxidase activity, whereas carmustine showed an insignificant effect. These observations suggest that glutathione and glutathione reductase activity contribute only partially to the total peroxidase activity of the sperm extract. While the specific role of PRDX2 in the total peroxidase activity of sperm extract is still an open question, the present study for the first time (to our knowledge) shows the presence of PRDX2 in mammalian spermatozoa. Peroxidase activity in sperm extracts is not due to the glutathione system and therefore possibly involves PRDX2 and other peroxiredoxins.


Subject(s)
Peroxidases/metabolism , Peroxiredoxins/metabolism , Spermatids/enzymology , Animals , Antineoplastic Agents, Alkylating , Carmustine , Cytoplasm/metabolism , Hydrogen Peroxide/metabolism , Immunohistochemistry , Male , Mice , NADP/metabolism , Peroxiredoxins/chemistry , Solubility , Sperm Head/enzymology , Swine , Testis/enzymology
10.
Mol Reprod Dev ; 76(5): 490-500, 2009 May.
Article in English | MEDLINE | ID: mdl-19090011

ABSTRACT

The following experiments compared the efficiency of three fusion/activation protocols following somatic cell nuclear transfer (SCNT) with porcine somatic cells transfected with enhanced green fluorescent protein driven by the chicken beta-actin/rabbit beta-globin hybrid promoter (pCAGG-EGFP). The three protocols included electrical fusion/activation (NT1), electrical fusion/activation followed by treatment with a reversible proteasomal inhibitor MG132 (NT2) and electrical fusion in low Ca(2+) followed by chemical activation with thimerosal/dithiothreitol (NT3). Data were collected at Days 6, 12, 14, 30, and 114 of gestation. Fusion rates, blastocyst-stage mean cell numbers, recovery rates, and pregnancy rates were calculated and compared between protocols. Fusion rates were significantly higher for NT1 and NT2 compared to NT3 (P < 0.05). There was no significant difference in mean nuclear number. Pregnancy rate for NT2 was 100% (n = 19) at all stages collected and was significantly higher than NT1 (71.4%, n = 28; P < 0.05), but was not significantly higher than NT3 (82.6%, n = 23; P < 0.15). Recovery rates were calculated based on the number of embryos, conceptuses, fetuses, or piglets present at the time of collection, divided by the number of embryos transferred to the recipient gilts. Recovery rates between the three groups were not significantly different at any of the stages collected (P > 0.05). All fusion/activation treatments produced live, pCAGG-EGFP positive piglets from SCNT. Treatment with MG132 after fusion/activation of reconstructed porcine embryos was the most effective method when comparing the overall pregnancy rates. The beneficial effect of NT2 protocol may be due to the stimulation of proteasomes that infiltrate donor cell nucleus shortly after nuclear transfer.


Subject(s)
Cloning, Organism/methods , Nuclear Transfer Techniques , Oocytes/physiology , Swine/physiology , Analysis of Variance , Animals , Blastocyst , Calcium/pharmacology , Cell Nucleus , Cellular Reprogramming , Electric Stimulation , Embryo Transfer , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Leupeptins/pharmacology , Microscopy, Fluorescence , Oocytes/drug effects , Pregnancy , Pregnancy Rate , Swine/genetics , Thimerosal/pharmacology , Zygote/metabolism
11.
Biol Reprod ; 78(1): 115-26, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17942798

ABSTRACT

The resumption of oocyte meiosis in mammals encompasses the landmark event of oocyte germinal vesicle (GV) breakdown (GVBD), accompanied by the modification of cell-to-cell communication and adhesion between the oocyte and surrounding cumulus cells. The concomitant cumulus expansion relies on microfilament-cytoskeletal remodeling and extracellular matrix (ECM) deposition. We hypothesized that this multifaceted remodeling event requires substrate-specific proteolysis by the ubiquitin-proteasome pathway (UPP). We evaluated meiotic progression, cytoskeletal dynamics, and the production of cumulus ECM in porcine cumulus-oocyte complexes (COCs) cultured with or without 10-200 microM MG132, a specific proteasomal inhibitor, for the first 22 h of in vitro maturation, followed by 22 h of culture with or without MG132. Treatment with 10 microM MG132 arrested 28.4% of oocytes in GV stage (vs. 1.3% in control), 43.1% in prometaphase I, and 16.2% in metaphase I, whereas 83.7% of control ova reached metaphase II (0% of MG132 reached metaphase II). The proportion of GV-stage ova increased progressively to >90% with increased concentration of MG132 (20-200 microM). Furthermore, MG132 blocked the extrusion of the first polar body and degradation of F-actin-rich transzonal projections (TZP) interconnecting cumulus cells with the oocyte. The microfilament disruptor cytochalasin E (CE) prevented cumulus expansion but accelerated the breakdown of TZPs. Ova treated with a combination of 10 microM MG132 and 10 microM CE underwent GVBD, despite the inhibition of proteasomal activity. However, 90.0% of cumulus-free ova treated with 10 microM MG132 remained in GV stage, compared with 16.7% GV ova in control. Cumulus expansion, retention of hyaluronic acid, and the deposition of cumulus ECM relying on the covalent transfer of heavy chains of inter-alpha trypsin inhibitor (IalphaI) were also inhibited by MG132. Cumulus expansion in control COCs was accompanied by the degradation of ubiquitin-C-terminal hydrolase L3, an important regulator of UPP. RAC1, a UPP-controlled regulator of actin polymerization was maintained at steady levels throughout cumulus expansion. We conclude that proteasomal proteolysis has multiple functions in the progression of oocyte meiosis beyond GV and metaphase I stage, polar body extrusion, and cumulus expansion.


Subject(s)
Cumulus Cells/physiology , Meiosis/physiology , Oocytes/physiology , Proteasome Endopeptidase Complex/metabolism , Swine , Actin Cytoskeleton/metabolism , Animals , Cells, Cultured , Cumulus Cells/cytology , Cumulus Cells/drug effects , Cytoplasm/metabolism , Cytoskeleton/drug effects , Dose-Response Relationship, Drug , Extracellular Matrix , Female , Gonadotropins/pharmacology , Leupeptins/pharmacology , Meiosis/drug effects , Oocytes/cytology , Oocytes/drug effects , Ovarian Follicle
12.
J Cell Physiol ; 215(3): 684-96, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18064599

ABSTRACT

Posttranslational modification by ubiquitination marks defective or outlived intracellular proteins for proteolytic degradation by the 26S proteasome. The ATP-dependent, covalent ligation and formation of polyubiquitin chains on substrate proteins requires the presence and activity of a set of ubiquitin activating and conjugating enzymes. While protein ubiquitination typically occurs in the cell cytosol or nucleus, defective mammalian spermatozoa become ubiquitinated on their surface during post-testicular sperm maturation in the epididymis, suggesting an active molecular mechanism for sperm quality control. Consequently, we hypothesized that the bioactive constituents of ubiquitin-proteasome pathway were secreted in the mammalian epididymal fluid (EF) and capable of ubiquitinating extrinsic substrates. Western blotting indeed detected the presence of the ubiquitin-activating enzyme E1 and presumed E1-ubiquitin thiol-ester intermediates, ubiquitin-carrier enzyme E2 and presumed E2-ubiquitin thiol-ester intermediates and the ubiquitin C-terminal hydrolase PGP 9.5/UCHL1 in the isolated bovine EF. Thiol-ester assays utilizing recombinant ubiquitin-activating and ubiquitin-conjugating enzymes, biotinylated substrates, and isolated bovine EF confirmed the activity of the ubiquitin activating and conjugating enzymes within EF. Ubiquitinated proteins were found to be enriched in the defective bull sperm fraction and appropriate proteasomal deubiquitinating and proteolytic activities were measured in the isolated EF by specific fluorescent substrates. The apocrine secretion of cytosolic proteins was visualized in transgenic mice and rats expressing the enhanced green fluorescent protein (eGFP) under the direction of ubiquitin-C promoter. Accumulation of eGFP, ubiquitin and proteasomes was detected in the apical blebs, the apocrine secretion sites of the caput epididymal epithelia of both the rat and mouse epididymal epithelium, although region-specific differences exist. Secretion of eGFP and proteasomes continued during the prolonged culture of the isolated rat epididymal epithelial cells in vitro. This study provides evidence that the activity of the ubiquitin system is not limited to the intracellular environment, contributing to a greater understanding of the sperm maturation process during epididymal passage.


Subject(s)
Epididymis/metabolism , Extracellular Space/metabolism , Mammals/metabolism , Ubiquitination , Animals , Animals, Genetically Modified , Biotinylation , Body Fluids/enzymology , Cattle , Cell Separation , Cells, Cultured , Culture Media , Cytoplasmic Structures/metabolism , Endopeptidases/metabolism , Epididymis/cytology , Epididymis/enzymology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fluorescent Dyes/metabolism , Green Fluorescent Proteins/metabolism , Male , Muramidase/metabolism , Promoter Regions, Genetic/genetics , Proteasome Endopeptidase Complex/metabolism , Rats , Sulfhydryl Compounds/metabolism , Ubiquitin C/genetics , Ubiquitin-Protein Ligase Complexes/metabolism
13.
Biol Reprod ; 77(5): 780-93, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17671268

ABSTRACT

The 26S proteasome, which is a multi-subunit protease with specificity for substrate proteins that are postranslationally modified by ubiquitination, has been implicated in acrosomal function and sperm-zona pellucida (ZP) penetration during mammalian fertilization. Ubiquitin C-terminal hydrolases (UCHs) are responsible for the removal of polyubiquitin chains during substrate priming for proteasomal proteolysis. The inhibition of deubiquitination increases the rate of proteasomal proteolysis. Consequently, we have hypothesized that inhibition of sperm acrosome-borne UCHs increases the rate of sperm-ZP penetration and polyspermy during porcine in vitro fertilization (IVF). Ubiquitin aldehyde (UA), which is a specific nonpermeating UCH inhibitor, significantly (P < 0.05) increased polyspermy during porcine IVF and reduced (P < 0.05) UCH enzymatic activity measured in motile boar spermatozoa using a specific fluorometric UCH substrate, ubiquitin-AMC. Antibodies against two closely related UCHs, UCHL1 and UCHL3, detected these UCHs in the oocyte cortex and on the sperm acrosome, respectively, and increased the rate of polyspermy during IVF, consistent with the UA-induced polyspermy surge. In the oocyte, UCHL3 was primarily associated with the meiotic spindle. Sperm-borne UCHL3 was localized to the acrosomal surface and coimmunoprecipitated with a peripheral acrosomal membrane protein, spermadhesin AQN1. Recombinant UCHs, UCHL3, and isopeptidase T reduced polyspermy when added to the fertilization medium. UCHL1 was detected in the oocyte cortex but not on the sperm surface, and was partially degraded 6-8 h after fertilization. Enucleated oocyte-somatic cell electrofusion caused polarized redistribution of cortical UCHL1. We conclude that sperm-acrosomal UCHs are involved in sperm-ZP interactions and antipolyspermy defense. Modulation of UCH activity could facilitate the management of polyspermy during IVF and provide insights into male infertility.


Subject(s)
Acrosome Reaction , Spermatozoa/enzymology , Ubiquitin Thiolesterase/metabolism , Acrosome/enzymology , Acrosome Reaction/drug effects , Amino Acid Sequence , Animals , Cysteine Endopeptidases/metabolism , Female , Fertilization , Fertilization in Vitro , Male , Membrane Proteins/metabolism , Molecular Sequence Data , Oocytes/enzymology , Sus scrofa , Ubiquitin Thiolesterase/analysis , Ubiquitin Thiolesterase/antagonists & inhibitors , Zona Pellucida/physiology
14.
Soc Reprod Fertil Suppl ; 63: 385-408, 2007.
Article in English | MEDLINE | ID: mdl-17566286

ABSTRACT

Despite years of research work, biologists remain divided over the issue of zona pellucida function during fertilization and the mode of sperm-ZP penetration. The present review examines the emerging evidence for the participation of ubiquitin-proteasome pathway in the process of sperm-ZP penetration generated in the last five years in species of mammals, ascidians and invertebrates. The 26S proteasome, a multi-subunit protease, selectively recognizes, and degrades, egg coat substrate proteins tagged by a covalent ligation of a small, multimeric protein, ubiquitin. Our in vitro work with pig gametes indicates that the sperm-borne 26S proteasomes selectively degrade an ubiquitinated ZP (glyco)protein during fertilization. We suggest that one or more of the ZP proteins are ubiquitinated, and proteasomes associated with the inner acrosomal membrane, are exposed as a result of acrosomal exocytosis. Sperm-ZP penetration may involve the ZP-deubiquitination, with several proteasomal subunits becoming phosphorylated. Polyubiquitin chain recognition activities associated with the sperm acrosomal proteasome could also contribute to anti-polyspermy control after sperm-egg fusion. Here, we bring together the relevant recent data on the mechanism of sperm-ZP penetration in mammals. Such observations could possibly lead to the development of novel non-hormonal contraceptives, improvement of infertility diagnostics and optimization of assisted reproduction.


Subject(s)
Mammals/physiology , Proteasome Endopeptidase Complex/metabolism , Sperm Head/enzymology , Sperm-Ovum Interactions/physiology , Zona Pellucida/metabolism , Animals , Female , Humans , Male , Microscopy, Electron, Transmission , Models, Animal , Sperm Head/ultrastructure , Ubiquitin/metabolism , Zona Pellucida/ultrastructure
15.
J Biol Chem ; 282(16): 12164-75, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17289678

ABSTRACT

We report a novel alkaline extractable protein of the sperm head that exclusively resides in the post-acrosomal sheath region of the perinuclear theca (PT) and is expressed and assembled in elongating spermatids. It is a protein that shares sequence homology to the N-terminal half of WW domain-binding protein 2, while the C-terminal half is unique and rich in proline. A functional PPXY consensus binding site for group-I WW domain-containing proteins, and numerous unique repeating motifs, YGXPPXG, are identified in the proline-rich region. Considering these molecular characteristics, we designated this protein PAWP for postacrosomal sheath WW domain-binding protein. Microinjection of recombinant PAWP or alkaline PT extract into metaphase II-arrested porcine, bovine, macaque, and Xenopus oocytes induced a high rate of pronuclear formation, which was prevented by co-injection of a competitive PPXY motif containing peptide derived from PAWP but not by co-injection of the point-mutated peptide. Intracytoplasmic sperm injection (ICSI) of porcine oocytes combined with co-injection of the competitive PPXY peptide or an anti-recombinant PAWP antiserum prevented pronuclear formation and arrested fertilization. Conversely, co-injection of the modified PPXY peptide, when the tyrosine residue of PPXY was either phosphorylated or substituted with phenylalanine, did not prevent ICSI-induced fertilization. This study uncovers a group I WW domain module signal transduction event within the fertilized egg that appears compulsory for meiotic resumption and pronuclear development during egg activation and provides compelling evidence that a PPXY motif of sperm-contributed PAWP can trigger these events.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Nucleus/metabolism , Fertilization , Meiosis , Seminal Plasma Proteins/genetics , Seminal Plasma Proteins/physiology , Spermatozoa/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/biosynthesis , Cattle , Female , Fertilization in Vitro , Macaca , Male , Molecular Sequence Data , Seminal Plasma Proteins/biosynthesis , Sequence Homology, Amino Acid , Swine , Xenopus
16.
Reproduction ; 129(3): 269-82, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15749954

ABSTRACT

Major vault protein (MVP), also called lung resistance-related protein is a ribonucleoprotein comprising a major part (>70%) of the vault particle. The function of vault particle is not known, although it appears to be involved in multi-drug resistance and cellular signaling. Here we show that MVP is expressed in mammalian, porcine, and human ova and in the porcine preimplantation embryo. MVP was identified by matrix-assisted laser-desorption ionization-time-of-flight (MALDI-TOF) peptide sequencing and Western blotting as a protein accumulating in porcine zygotes cultured in the presence of specific proteasomal inhibitor MG132. MVP also accumulated in poor-quality human oocytes donated by infertile couples and porcine embryos that failed to develop normally after in vitro fertilization or somatic cell nuclear transfer. Normal porcine oocytes and embryos at various stages of preimplantation development showed mostly cytoplasmic labeling, with increased accumulation of vault particles around large cytoplasmic lipid inclusions and membrane vesicles. Occasionally, MVP was associated with the nuclear envelope and nucleolus precursor bodies. Nucleotide sequences with a high degree of homology to human MVP gene sequence were identified in porcine oocyte and endometrial cell cDNA libraries. We interpret these data as the evidence for the expression and ubiquitin-proteasome-dependent turnover of MVP in the mammalian ovum. Similar to carcinoma cells, MVP could fulfill a cell-protecting function during early embryonic development.


Subject(s)
Mammals/metabolism , Oocytes/metabolism , Vault Ribonucleoprotein Particles/metabolism , Zygote/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western/methods , Cell Culture Techniques , Electrophoresis, Polyacrylamide Gel , Female , Fertilization in Vitro , Fluorescent Antibody Technique , Humans , Mice , Molecular Sequence Data , Oocytes/chemistry , Oogenesis , Sequence Analysis, Protein , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Swine
17.
Biol Reprod ; 72(1): 2-13, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15385423

ABSTRACT

Animal spermatids and primary oocytes initially have typical centrosomes comprising pairs of centrioles and pericentriolar fibrous centrosomal proteins. These somatic cell-like centrosomes are partially or completely degenerated during gametogenesis. Centrosome reduction during spermiogenesis comprises attenuation of microtubule nucleation function, loss of pericentriolar material, and centriole degeneration. Centrosome reduction during oogenesis is due to complete degeneration of centrioles, which leads to dispersal of the pericentriolar centrosomal proteins, loss of replicating capacity of the spindle poles, and switching to acentrosomal mode of spindle organization. Oocyte centrosome reduction plays an important role in preventing parthenogenetic embryogenesis and balancing centrosome number in the embryonic cells.


Subject(s)
Centrosome/physiology , Oogenesis/physiology , Spermatogenesis/physiology , Animals , Centrosome/chemistry , Female , Humans , Male , Meiosis , Oocytes/physiology , Spermatozoa/physiology , Spindle Apparatus/physiology
18.
Biol Reprod ; 71(5): 1625-37, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15253927

ABSTRACT

The ubiquitin-proteasome pathway has been implicated in the penetration of ascidian vitelline envelope by the fertilizing spermatozoon (Sawada et al., Proc Natl Acad Sci U S A 2002; 99:1223-1228). The present study provides experimental evidence demonstrating proteasome involvement in the penetration of mammalian zona pellucida (ZP). Using porcine in vitro fertilization as a model, penetration of ZP was completely inhibited by specific proteasomal inhibitors MG-132 and lactacystin. Three commercial rabbit sera recognizing 20S proteasomal core subunits beta-1i, beta-2i, alpha-6, and beta-5 completely blocked fertilization at a very low concentration (i.e., diluted 1/2000 to 1/8000 in fertilization medium). Neither proteasome inhibitors nor antibodies had any effects on sperm-ZP binding and acrosome exocytosis in zona-enclosed oocytes or on fertilization rates in zona-free oocytes, which were highly polyspermic. Consistent with a possible role of ubiquitin-proteasome pathway in ZP penetration, ubiquitin and various alpha and beta type proteasomal subunits were detected in boar sperm acrosome by specific antibodies, immunoprecipitated and microsequenced by MALDI-TOF from boar sperm extracts. Antiubiquitin-immunoreactive substrates were detected on the outer face of ZP by epifluorescence microscopy. This study therefore provides strong evidence implicating the ubiquitin-proteasome pathway in mammalian fertilization and zona penetration. This finding opens a new line of acrosome/ZP research because further studies of the sperm acrosomal proteasome can provide new tools for the management of polyspermia during in vitro fertilization and identify new targets for contraceptive development.


Subject(s)
Acetylcysteine/analogs & derivatives , Fertilization/physiology , Proteasome Endopeptidase Complex/physiology , Sperm-Ovum Interactions/physiology , Zona Pellucida/physiology , Acetylcysteine/pharmacology , Acrosome/metabolism , Acrosome Reaction/drug effects , Animals , Exocytosis/physiology , Female , Fertilization in Vitro , Immune Sera/immunology , Immune Sera/pharmacology , Leupeptins/pharmacology , Male , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Sperm-Ovum Interactions/drug effects , Spermatozoa/metabolism , Swine , Ubiquitin/metabolism , Zona Pellucida/metabolism
19.
Microsc Res Tech ; 61(4): 362-78, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12811742

ABSTRACT

Perinuclear theca (PT) is the cytoskeletal coat of mammalian sperm nucleus that is removed from the sperm head at fertilization. PT harbors the sperm borne, oocyte-activating factor (SOAF), a yet-to-be-characterized substance responsible for triggering the signaling cascade of oocyte activation, thought to be dependent on intra-oocyte calcium release. The present article reviews the current knowledge on the biogenesis and molecular composition of sperm PT. Possible functions of sperm PT during natural and assisted fertilization, and in the initiation of embryonic development are discussed. Furthermore, evidence is provided that SOAF is transferred from the sperm PT to oocyte cytoplasm through the internalization and rapid solubilization of the post-acrosomal PT. It is shown that during natural fertilization the sperm PT dissolves in the oocyte cytoplasm concomitantly with sperm nuclear decondensation and the initiation of pronuclear development. SOAF activity is preserved in the differentially extracted sperm heads only if the integrity of PT is maintained. After intracytoplasmic sperm injection (ICSI), activation occurs only in those oocytes in which the injected spermatozoon displays complete or partial dissolution of PT. In the latter case, the residual PT of the sub-acrosomal and/or post-acrosomal sperm region may persist on the apical surface of the sperm nucleus/male pronucleus and may cause a delay or arrest of zygotic development. We propose that the sperm PT harbors SOAF in the post-acrosomal sheath, as this is the first part of the sperm cytosol to enter the oocyte cytoplasm and its disassembly appears sufficient to initiate the early events of oocyte activation. Dissolution of the sub-acrosomal part of the PT, on the other hand, appears necessary to insure complete DNA decondensation in the internalized sperm nucleus and initiate DNA synthesis of both pronuclei. The release of the SOAF from the sperm head into oocyte cytoplasm at fertilization ultimately leads to the activation of oocyte mechanism including the completion of the meiotic cell cycle, pronuclear development and anti-polyspermy defense.


Subject(s)
Fertilization/physiology , Oocytes/growth & development , Oocytes/physiology , Sperm Head/physiology , Animals , Cattle , Female , Male , Microscopy, Electron , Oocytes/ultrastructure , Reproduction/physiology , Sperm Head/ultrastructure , Sperm-Ovum Interactions , Spermatozoa/physiology , Spermatozoa/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...