Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Cell Tissue Res ; 387(1): 159-171, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34762184

RESUMO

Low fertility is the single most important factor limiting livestock reproductive performance, adversely affecting the cattle industry and causing millions of dollars of economic loss. In the livestock industry, male fertility is of crucial importance for the reproductive performance of livestock. However, there is a lack of reliable biomarkers to predict bull fertility in artificial insemination service. The objective of this study was to identify sperm proteins as biomarkers for bull fertility. To discover candidate sperm quality biomarkers, sperm proteome profiling was conducted in extreme high- and extreme low-fertile bulls selected from a pool of 1000 AI sires with varied fertility. Thirty-two differentially expressed proteins were identified. Among them, high levels of sperm outer dense fiber of sperm tails 2 (ODF2) and post-acrosomal assembly of sperm head protein (PAWP/WBP2NL) represented the most extreme differences in quantity between high- and low-fertility bulls. Protein immunodetection and flow cytometry used to validate these putative fertility markers in a combined cohort of 154 AI sires. Both ODF2 and PAWP correlated significantly with fertility. In conclusion, ODF2 and PAWP can be used to assess semen quality and predict sire fertility.


Assuntos
Biomarcadores/metabolismo , Fertilidade/fisiologia , Análise do Sêmen/veterinária , Espermatozoides/fisiologia , Animais , Bovinos , Masculino
2.
Adv Exp Med Biol ; 1358: 345-367, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35641877

RESUMO

Structural and regulatory requirements of mammalian spermatozoa in both development and function make them extremely unique cells. Looking at the complexity of spermatozoon structure and its requirements for both motility and quick breakdown within the post-fertilization environment, as well as its functional needs as an extremely streamlined cell with high energy requirements, demonstrate the high importance of oxidative-reductive processes. The oxidative state of the testis and epididymis during sperm development and maturation highly influences sperm structure, with a high dependence on disulfide bond formation, facilitated by thiol mediated processes. However, once functionally active, sperm transition to a new high-risk functional paradigm requiring low levels of reactive oxygen species (ROS) while also being highly susceptible to oxidative damage due to the high proportion of polyunsaturated fatty acids within the lipid bilayer of the plasmalemma and the lack of cytosolic antioxidant defenses. This chapter highlights how glutathione and thioredoxin systems mediate the oxidative environment of the male reproductive tract and facilitate the successful development, maturation and function of mammalian spermatozoa.


Assuntos
Maturação do Esperma , Espermatozoides , Animais , Fertilidade , Fertilização , Masculino , Mamíferos , Oxirredução , Maturação do Esperma/fisiologia , Espermatozoides/metabolismo
3.
Int J Mol Sci ; 22(15)2021 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-34360885

RESUMO

The perinuclear theca (PT) of the eutherian sperm head is a cytoskeletal-like structure that houses proteins involved in important cellular processes during spermiogenesis and fertilization. Building upon our novel discovery of non-nuclear histones in the bovine PT, we sought to investigate whether this PT localization was a conserved feature of eutherian sperm. Employing cell fractionation, immunodetection, mass spectrometry, qPCR, and intracytoplasmic sperm injections (ICSI), we examined the localization, developmental origin, and functional potential of histones from the murid PT. Immunodetection localized histones to the post-acrosomal sheath (PAS) and the perforatorium (PERF) of the PT but showed an absence in the sperm nucleus. MS/MS analysis of selectively extracted PT histones indicated that predominately core histones (i.e., H3, H3.3, H2B, H2A, H2AX, and H4) populate the murid PT. These core histones appear to be de novo-synthesized in round spermatids and assembled via the manchette during spermatid elongation. Mouse ICSI results suggest that early embryonic development is delayed in the absence of PT-derived core histones. Here, we provide evidence that core histones are de novo-synthesized prior to PT assembly and deposited in PT sub-compartments for subsequent involvement in chromatin remodeling of the male pronucleus post-fertilization.


Assuntos
Histonas/biossíntese , Cabeça do Espermatozoide/metabolismo , Espermátides/metabolismo , Espermatogênese/fisiologia , Animais , Núcleo Celular/metabolismo , Cromatografia Líquida/métodos , Feminino , Fertilização/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Injeções de Esperma Intracitoplásmicas , Espectrometria de Massas em Tandem/métodos
4.
Int J Mol Sci ; 21(6)2020 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-32204514

RESUMO

Building on our recent discovery of the zinc signature phenomenon present in boar, bull, and human spermatozoa, we have further characterized the role of zinc ions in the spermatozoa's pathway to fertilization. In boar, the zinc signature differed between the three major boar ejaculate fractions, the initial pre-rich, the sperm-rich, and the post-sperm-rich fraction. These differences set in the sperm ejaculatory sequence establish two major sperm cohorts with marked differences in their sperm capacitation progress. On the subcellular level, we show that the capacitation-induced Zn-ion efflux allows for sperm release from oviductal glycans as analyzed with the oviductal epithelium mimicking glycan binding assay. Sperm zinc efflux also activates zinc-containing enzymes and proteases involved in sperm penetration of the zona pellucida, such as the inner acrosomal membrane matrix metalloproteinase 2 (MMP2). Both MMP2 and the 26S proteasome showed severely reduced activity in the presence of zinc ions, through studies using by gel zymography and the fluorogenic substrates, respectively. In the context of the fertilization-induced oocyte zinc spark and the ensuing oocyte-issued polyspermy-blocking zinc shield, the inhibitory effect of zinc on sperm-borne enzymes may contribute to the fast block of polyspermy. Altogether, our findings establish a new paradigm on the role of zinc ions in sperm function and pave the way for the optimization of animal semen analysis, artificial insemination (AI), and human male-factor infertility diagnostics.


Assuntos
Fertilização/fisiologia , Oviductos/metabolismo , Capacitação Espermática/fisiologia , Espermatozoides/metabolismo , Zinco/metabolismo , Zona Pelúcida/metabolismo , Acrossomo/metabolismo , Animais , Bovinos , Feminino , Humanos , Transporte de Íons , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Oócitos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Análise do Sêmen/métodos , Suínos
5.
Biol Reprod ; 100(6): 1461-1472, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30939204

RESUMO

The perinuclear theca (PT) is a cytosolic protein capsule that surrounds the nucleus of eutherian spermatozoa. Compositionally, it is divided into two regions: the subacrosomal layer (SAL) and the postacrosomal sheath (PAS). In falciform spermatozoa, a third region of the PT emerges that extends beyond the nuclear apex called the perforatorium. The formation of the SAL and PAS differs, with the former assembling early in spermiogenesis concomitant with acrosome formation, and the latter dependent on manchette descent during spermatid elongation. The perforatorium also forms during the elongation phase of spermiogenesis, suggesting that like the PAS, its assembly is facilitated by the manchette. The temporal similarity in biogenesis between the PAS and perforatorium led us to compare their molecular composition using cell fractionation and immunodetection techniques. Although the perforatorium is predominantly composed of its endemic protein FABP9/PERF15, immunolocalization indicates that it also shares proteins with the PAS. These include WBP2NL/PAWP, WBP2, GSTO2, and core histones, which have been implicated in early fertilization and zygotic events. The compositional homogeny between the PAS and perforatorium supports our observation that their development is linked. Immunocytochemistry indicates that both PAS and perforatorial biogenesis depend on the transport and deposition of cytosolic proteins by the microtubular manchette. Proteins translocated from the manchette pass ventrally along the spermatid head into the apical perforatorial space prior to PAS deposition in the wake of manchette descent. Our findings demonstrate that the perforatorium and PAS share a mechanism of developmental assembly and thereby contain common proteins that facilitate fertilization.


Assuntos
Acrossomo , Proteínas/metabolismo , Cabeça do Espermatozoide , Espermatogênese/fisiologia , Acrossomo/metabolismo , Acrossomo/ultraestrutura , Animais , Bovinos , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Humanos , Masculino , Proteínas/análise , Ratos , Ratos Sprague-Dawley , Análise do Sêmen , Cabeça do Espermatozoide/metabolismo , Cabeça do Espermatozoide/ultraestrutura , Espermatozoides/citologia , Espermatozoides/metabolismo , Espermatozoides/ultraestrutura , Suínos
6.
Biol Reprod ; 101(2): 368-376, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31087045

RESUMO

The postacrosomal sheath (PAS) of the perinuclear theca (PT) is the first compartment of the sperm head to solubilize into the ooplasm upon sperm-oocyte fusion, implicating its constituents in zygotic development. This study investigates the role of one such constituent, glutathione-S-transferase omega 2 (GSTO2), an oxidative-reductive enzyme found in the PAS and perforatorial regions of the PT. GSTO2 uses the conjugation of reduced glutathione, an electron donor shown to be compulsory in sperm disassembly within the ooplasm. The proximity of GSTO2 to the condensed sperm nucleus led us to hypothesize that this enzyme may facilitate nuclear decondensation by reducing disulfide bonds before the recruitment of GSTO enzymes from within the ooplasm. To test this hypothesis, we utilized a cell permeable isozyme-specific inhibitor, which fluoresces when bound to the active site of GSTO2, to functionally inhibit spermatozoa before performing intracytoplasmic sperm injections (ICSI) in mice. The technique allowed for targeted inhibition of solely PT-residing GSTO2, as all that is required for complete zygotic development is the injection of the mouse spermatozoon head. ICSI showed that inhibition of PT-anchored GSTO2 caused a delay in sperm nuclear decondensation, and further resulted in untimely embryo cleavage, and an increase in fragmentation beginning at the morula stage. The confounding effects of these developmental delays ultimately resulted in decreased blastocyst formation. This study implicates PT-anchored GSTO2 as an important facilitator of nuclear decondensation and reinforces the notion that the PAS-PT is a critical sperm compartment harboring molecules that facilitate zygotic development.


Assuntos
Glutationa Transferase/metabolismo , Cabeça do Espermatozoide/fisiologia , Espermatozoides/enzimologia , Sequência de Aminoácidos , Animais , Feminino , Glutationa Transferase/química , Glutationa Transferase/genética , Masculino , Camundongos , Injeções de Esperma Intracitoplásmicas/métodos , Interações Espermatozoide-Óvulo/fisiologia
7.
Biol Reprod ; 99(6): 1171-1183, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30010725

RESUMO

The sperm-borne oocyte-activating factor (SOAF) resides in the sperm perinuclear theca (PT). A consensus has been reached that SOAF most likely resides in the postacrosomal sheath (PAS), which is the first region of the PT to solubilize upon sperm-oocyte fusion. There are two SOAF candidates under consideration: PLCZ1 and WBP2NL. A mouse gene germline ablation of the latter showed that mice remain fertile with no observable phenotype despite the fact that a competitive inhibitor of WBP2NL, derived from its PPXY motif, blocks oocyte activation when coinjected with WBP2NL or spermatozoa. This suggested that the ortholog of WBP2NL, WBP2, containing the same domain and motifs associated with WBP2NL function, might compensate for its deficiency in oocyte activation. Our objectives were to examine whether WBP2 meets the developmental criteria established for SOAF and whether it has oocyte-activating potential. Immunoblotting detected WBP2 in mice testis and sperm and immunofluorescence localized WBP2 to the PAS and perforatorium of the PT. Immunohistochemistry of the testes revealed that WBP2 reactivity was highest in round spermatids and immunofluorescence detected WBP2 in the cytoplasmic lobe of elongating spermatids and colocalized it with the microtubular manchette during PT assembly. Microinjection of the recombinant forms of WBP2 and WBP2NL into metaphase II mouse oocytes resulted in comparable rates of oocyte activation. This study shows that WBP2 shares a similar testicular developmental pattern and location with WBP2NL and a shared ability to activate the oocyte, supporting its consideration as a mouse SOAF component that can compensate for a WBP2NL.


Assuntos
Proteínas de Transporte/metabolismo , Oócitos/fisiologia , Proteínas de Plasma Seminal/metabolismo , Animais , Anticorpos , Proteínas de Transporte/genética , Bovinos , Humanos , Masculino , Camundongos , Transporte Proteico , Proteínas de Plasma Seminal/genética , Especificidade da Espécie , Transativadores
8.
Biol Reprod ; 97(4): 612-621, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-29036365

RESUMO

The perinuclear theca (PT) is a condensed, nonionic detergent resistant cytosolic protein layer encapsulating the sperm head nucleus. It can be divided into two regions: the subacrosomal layer, whose proteins are involved in acrosomal assembly during spermiogenesis, and the postacrosomal sheath (PAS), whose proteins are implicated in sperm-oocyte interactions during fertilization. In continuation of our proteomic analysis of the PT, we have isolated two prominent PT-derived proteins of 28 and 31 kDa from demembranated bovine sperm head fractions. These proteins were identified by mass spectrometry as isoforms of glutathione-s-transferase omega 2 (GSTO2). Immunoblots probed with anti-GSTO2 antibodies confirmed the presence of the GSTO2 isoforms in these fractions while fluorescent immunocytochemistry localized the isoforms to the PAS region of the bull, boar, and murid PT. In addition to the PAS labeling of GSTO2, the performatorium of murid spermatozoa was also labeled. Immunohistochemistry of rat testes revealed that GSTO2 was expressed in the third phase of spermatogenesis (i.e., spermiogenesis) and assembled in the PAS and perforatorial regions of late elongating spermatids. Fluorescent immunocytochemistry performed on murine testis cells co-localized GSTO2 and tubulin on the transient microtubular-manchette of elongating spermatids. These findings imply that GSTO2 is transported and deposited in the PAS region by the manchette, conforming to the pattern of assembly found with other PAS proteins. The late assembly of GSTO2 and its localization in the PAS suggests a role in regulating the oxidative and reductive state of covalently linked spermatid/sperm proteins, especially during the disassembly of the sperm accessory structures after fertilization.


Assuntos
Bovinos/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Glutationa Transferase/metabolismo , Cabeça do Espermatozoide/enzimologia , Cabeça do Espermatozoide/fisiologia , Animais , Glutationa Transferase/genética , Masculino , Isoformas de Proteínas , Especificidade da Espécie
9.
Biol Reprod ; 94(4): 88, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26935599

RESUMO

Spermatozoa must penetrate the outer investments of the oocyte, the cumulus oophorus and the zona pellucida (ZP), in order for fertilization to occur. This may require exposure of enzymes on the sperm's inner acrosomal membrane (IAM), one of which is matrix metalloproteinase (MMP) 2, to factors in oviductal fluid. Plasminogen is present in oviductal fluid and activates MMP2 in somatic tissues. The objectives of this study were: 1) to examine possible interactions between plasminogen and IAM-bound plasminogen activator receptor (SAMP14) and -MMP2, 2) to demonstrate plasminogen's presence in the extracellular environment at the site of fertilization, and 3) to provide evidence that plasminogen plays a role in fertilization. Zymographs of sonicated bull and rat sperm extracts incubated with plasmin and/or plasminogen (plasmin/ogen) showed acceleration of initiation of MMP2 activity in concentrations as low as 1 µg/ml. Immunohistochemical and immunofluorescence analysis of plasmin/ogen revealed its presence in the cytoplasm of mouse ovarian and oviductal oocytes, oviductal epithelium, around the ZP, and amongst the cumulus cells. We modified the standard in vitro fertilization (IVF) approach to more closely mimic natural fertilization by reducing sperm concentration during insemination by ∼100× and also comparing cumulus-intact and denuded oocytes. In mice, addition of plasminogen in IVF medium significantly improved fertilization, while MMP2 antibody significantly inhibited sperm penetration in these conditions. IVF improvement by plasminogen was blocked by SAMP14 antibody. Furthermore, MMP2 antibody inhibition was coincident with a failure by spermatozoa to disperse the cumulus oophorus. We provide evidence that plasminogen on its own and through an MMP2-related mechanism improves the ability of oocytes to be fertilized, and demonstrate its effect in sperm penetration of oocyte investments.


Assuntos
Fertilização in vitro/métodos , Metaloproteinase 2 da Matriz/metabolismo , Plasminogênio/metabolismo , Receptores de Superfície Celular/metabolismo , Acrossomo/metabolismo , Animais , Bovinos , Feminino , Fertilização , Masculino , Camundongos , Ratos
10.
FASEB J ; 28(10): 4434-40, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24970390

RESUMO

Mammalian zygotic development is initiated by sperm-mediated intracellular calcium oscillations, followed by activation of metaphase II-arrested oocytes. Sperm postacrosomal WW binding protein (PAWP) fulfils the criteria set for an oocyte-activating factor by inducing oocyte activation and being stored in the perinuclear theca, the sperm compartment whose content is first released into oocyte cytoplasm during fertilization. However, proof that PAWP initiates mammalian zygotic development relies on demonstration that it acts upstream of oocyte calcium oscillations. Here, we show that PAWP triggers calcium oscillations and pronuclear formation in human and mouse oocytes similar to what is observed during intracytoplasmic sperm injection (ICSI). Most important, sperm-induced calcium oscillations are blocked by coinjection of a competitive inhibitor, derived from the WWI domain-binding motif of PAWP, implying the requirement of sperm PAWP and an oocyte-derived WWI domain protein substrate of PAWP for successful fertilization. Sperm-delivered PAWP is, therefore, a unique protein with a nonredundant role during human and mouse fertilization, required to trigger zygotic development. Presented data confirm our previous findings in nonmammalian models and suggest potential applications of PAWP in the diagnosis and treatment of infertility.-


Assuntos
Sinalização do Cálcio , Proteínas de Transporte/farmacologia , Oócitos/efeitos dos fármacos , Proteínas de Plasma Seminal/farmacologia , Animais , Feminino , Humanos , Camundongos , Oócitos/metabolismo
11.
Hum Reprod ; 29(4): 683-96, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24549217

RESUMO

STUDY QUESTION: Does antimicrobial peptide, LL-37, inhibit sperm fertilizing ability? SUMMARY ANSWER: Our results indicate that LL-37 inhibits mouse and human sperm fertilizing ability. WHAT IS KNOWN ALREADY: LL-37, a cationic antimicrobial peptide, exerts its microbicidal effects through the disruption of microbial cytoplasmic membranes following its interaction with microbial surface anionic phospholipids. ALL-38 (an LL-37 close analogue: LL-37 + Ala at the N-terminus) is produced in the vagina 2-6 h post-intercourse from its precursor hCAP-18, a seminal plasma component. At this time, motile sperm have already swum into the uterine cavity, thus unexposed to ALL-38. Since sperm contain a substantial amount of acidic sulfogalactosylglycerolipid (SGG) on their surface, treatment of sperm with LL-37 may cause their membrane disruption in an analogous manner to that occurring on microbial membranes. STUDY DESIGN, SIZE AND DURATION: Mouse/human sperm treated (2-30 min) with LL-37 in a physiological concentration range (up to 10.8 µM) were assessed for SGG-dependent LL-37 binding, and parameters relevant to fertilizing ability, namely motility and intactness of the sperm acrosome and plasma membrane. Ability of mouse sperm to fertilize eggs in vitro was also evaluated. Each study was performed with greater than or equal to three different sperm samples. The efficacy of LL-37 to inhibit sperm fertilizing ability in vivo was determined in female mice (n = 26 each for LL-37 treatment and no treatment), using sperm retrieved from 26 males. PARTICIPANTS/MATERIALS, SETTING, METHODS: Human sperm samples were donated by fertile men. LL-37 was chemically synthesized and was biotinylated for sperm binding studies. Sperm motility was assessed by videomicroscopy and the acrosomal status by Coomassie blue staining of acrosome-intact mouse sperm or the exposure of CD46, an inner acrosomal membrane protein, of acrosome reacted human sperm. Sperm membrane permeabilization/disruption was assessed by the loss of hypo-osmotic swelling response, an incorporation of Sytox Green (a membrane impermeable fluorescent DNA dye), and electron microscopy. Mouse IVF was scored by the presence of two pronuclei in eggs 6 h post-insemination. Ability of mouse sperm to fertilize eggs in vivo was determined by the pregnancy outcome of female mice injected transcervically with sperm with or without LL-37. MAIN RESULTS AND THE ROLE OF CHANCE: Biotinylated LL-37 bound to both mouse and human sperm and the binding was partially dependent on sperm surface SGG. Mouse and human sperm became immotile and underwent a premature acrosome reaction upon treatment with LL-37 at 3.6 and 10.8 µM, respectively. The initial action of LL-37 on both mouse and human sperm appeared to be through permeabilization/disruption of sperm surface membranes evidenced by the loss of hypo-osmotic swelling response, Sytox Green staining and electron microscopy revealing ultrastructural damage. Mouse sperm treated with 3.6 µM LL-37 lost the ability to fertilize eggs both in vitro and in vivo. All 26 female mice inseminated with sperm and LL-37 did not become pregnant. No apparent damage to the reproductive tract was observed as revealed by histological characterization in LL-37-inseminated mice and these females resumed fecundity following mating with fertile males. LIMITATIONS, REASONS FOR CAUTION: Direct demonstration that LL-37 treated human sperm fail to fertilize eggs was limited by legal restrictions on obtaining human eggs for such use. WIDER IMPLICATIONS OF THE FINDINGS: Our results reveal selective inhibitory effects of LL-37 on sperm fertilizing ability in mice without apparent impairment to the female reproductive tract. LL-37 is therefore a promising candidate to be developed into a vaginal contraceptive with microbicidal activity. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by Grand Challenges Explorations grant from the Bill & Melinda Gates Foundation (OPP1024509), Canadian Institutes of Health Research (MOP119438 & CCI82413) and International Collaboration and Exchanges NSFC of China (No.30611120525). There are no competing interests to declare.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Anticoncepcionais , Fertilização/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Acrossomo/efeitos dos fármacos , Animais , Feminino , Humanos , Masculino , Camundongos , Motilidade dos Espermatozoides/efeitos dos fármacos , Interações Espermatozoide-Óvulo/efeitos dos fármacos , Catelicidinas
12.
Mol Reprod Dev ; 81(5): 436-49, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24488940

RESUMO

Post-acrosomal WW-domain binding protein (PAWP) is a signaling molecule located in the post-acrosomal sheath (PAS) of mammalian spermatozoa. We hypothesized that the proper integration of PAWP in the sperm PAS is reflective of bull-sperm quality and fertility. Cryopreserved semen samples from 298 sires of acceptable, but varied, fertility used in artificial insemination services were analyzed using immunofluorescence microscopy and flow cytometry for PAWP protein. In normal spermatozoa, PAWP fluorescence formed a regular band around the proximal PAS. Anomalies of PAWP labeling in defective spermatozoa were reflected in flow cytometry by varied intensities of PAWP-induced fluorescence. Distinct sperm phenotypes were also identified, including morphologically normal and some defective spermatozoa with moderate levels of PAWP; grossly defective spermatozoa with low/no PAWP; and defective spermatozoa with high PAWP. Analysis by ImageStream flow cytometry confirmed the prevalence of abnormal sperm phenotypes in the spermatozoa with abnormal PAWP content. Live/dead staining and video recording showed that some abnormal spermatozoa are viable and capable of progressive motility. Conventional flow-cytometric measurements of PAWP correlated significantly with semen quality and fertility parameters that reflect the sires' artificial insemination fertility, including secondary sperm morphology, conception rate, non-return rate, and residual value. A multiplex, flow-cytometric test detecting PAWP, aggresomes (ubiquitinated protein aggregates), and acrosomal integrity (peanut-agglutinin-lectin labeling) had a predictive value for conception rate, as demonstrated by step-wise regression analysis. We conclude that PAWP correlates with semen/fertility parameters used in the cattle artificial insemination industry, making PAWP a potential biomarker of bull fertility.


Assuntos
Fertilidade/fisiologia , Regulação da Expressão Gênica/fisiologia , Inseminação Artificial , Análise do Sêmen , Proteínas de Plasma Seminal/biossíntese , Espermatozoides/metabolismo , Animais , Bovinos , Masculino , Espermatozoides/citologia
13.
J Cell Physiol ; 228(8): 1629-37, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23359453

RESUMO

Prothymosin α (PTMA) is a highly acidic intrinsically unstructured protein. Its expression in male gonads is evolutionary conserved; in rat testis it is specifically localized in the cytoplasm of post-meiotic germ cells, in association with the developing acrosome system. In the present paper we investigated on PTMA localization inside the head of mammalian spermatozoa (SPZ). We chose a confocal approach to ascertain whether PTMA is expressed in the acrosome or in the perinuclear theca, two regions that are tightly linked and partially overlapped in the mature haploid cells. The obtained results showed that PTMA is specifically localized in the acrosome of rat epididymal SPZ; the same experimental approach evidenced, for the first time, PTMA presence in human ejaculated SPZ. A Western blot analysis on protein extracts from human sperm head fractions confirmed the confocal data and demonstrated that the peptide is specifically associated with the inner acrosomal membrane fraction. Finally, when the acrosome reaction was induced in vitro by progesterone treatment on both rat and human sperm, PTMA signal was retained in the apical region of reacted SPZ. In conclusion, this study confirms the conservation of PTMA distribution in vertebrate male gametes and strongly supports a role for this polypeptide in their physiology.


Assuntos
Acrossomo/metabolismo , Precursores de Proteínas/metabolismo , Espermatozoides/metabolismo , Timosina/análogos & derivados , Reação Acrossômica , Animais , Western Blotting , Compartimento Celular , Fracionamento Celular/métodos , Epididimo/citologia , Epididimo/metabolismo , Humanos , Masculino , Microscopia de Fluorescência , Peptídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Timosina/metabolismo
14.
Biol Reprod ; 88(3): 65, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23269664

RESUMO

Cystatin-related epididymal spermatogenic protein (CRES, also CST8) is expressed in both the testis and epididymis and found associated with spermatozoa. It appears as nonglycosylated (14 and 12 kDa) and glycosylated isoforms (19 and 17 kDa). The role of CRES remains enigmatic and is dependent on localization of its isoforms, which is the objective of this study. Our initial approach was to investigate testicular origins of these isoforms by immunohistochemistry and immunogold electron microscopy. We confirmed previous reports that CRES was expressed in the cytoplasm of elongating spermatids from step 8 to step 16. However, we noticed that the pattern of testicular expression was reminiscent of outer dense fiber (ODF) and fibrous sheath (FS) proteins. Western blot analysis of extracts of cauda epididymal sperm revealed a nonionic, detergent-insoluble 14-kDa CRES isoform. To further pinpoint and confirm CRES localization we separated sperm heads and tails and extracted the tails with progressively harsher protein solubilizing agents. Western blots of these sequential extracts, designed to progressively remove the mitochondrial sheath and the ODFs or FS, detected a CRES-immunoreactive 14-kDa band associated with the accessory fibers of the tail. Immunogold labeling was concentrated over growing ODFs in the testes and persisted in spermatozoa. This study discovers a CRES isoform that assembles as part of the ODFs during the elongation and maturation phases of spermiogenesis and is retained as a covalently bound component of the ODFs in spermatozoa.


Assuntos
Cistatinas/metabolismo , Cauda do Espermatozoide/metabolismo , Espermatogênese , Animais , Detergentes , Immunoblotting , Imuno-Histoquímica , Masculino , Camundongos , Microscopia Eletrônica , Cabeça do Espermatozoide/metabolismo , Cabeça do Espermatozoide/ultraestrutura , Cauda do Espermatozoide/ultraestrutura , Testículo/metabolismo
15.
Biol Reprod ; 89(2): 38, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23843230

RESUMO

Oviductin or OVGP1, also known as oviduct-specific glycoprotein, has been shown to enhance sperm capacitation in addition to its other beneficial effects on fertilization and early embryo development. We hypothesized that estrus stage-specific hamster oviductin (eHamOVGP1) can potentiate the enhancement of tyrosine phosphorylation of sperm proteins during capacitation. Immunofluorescent staining and confocal microscopy as well as immunocytochemistry and surface replica technique localized tyrosine-phosphorylated proteins to the equatorial segment and midpiece after incubation of hamster sperm in capacitation medium in the presence or absence of eHamOVGP1. Increase of tyrosine phosphorylation level in the equatorial segment occurred as early as 5 min after incubation in the presence of eHamOVGP1. Immunostaining for eHamOVGP1 further increased upon prolonged incubation of sperm in medium containing the glycoprotein. Regardless of the presence or absence of eHamOVGP1, phosphotyrosine expression was observed along the tail, particularly at the midpiece. Western blotting of NP40-extracted sperm proteins (25, 37, and 44 kDa) and NP40-non-extractable sperm proteins (70, 83, 90 kDa) showed increased immunolabeling intensity after 5, 60, 120, and 180 min of capacitation in the presence of eHamOVGP1. Mass spectrometric analysis identified several proteins of functions known to be involved in metabolic pathways responsible for enhancement of tyrosine phosphorylation in its presence. The present investigation provides evidence that eHamOVGP1 regulates the expression of protein tyrosine phosphorylation in sperm capacitated in vitro, further supporting an important role of the presence of OVGP1 in the oviductal milieu during the process of fertilization.


Assuntos
Serina Endopeptidases/metabolismo , Capacitação Espermática/fisiologia , Espermatozoides/metabolismo , Animais , Cricetinae , Feminino , Masculino , Fosforilação , Fosfotirosina/metabolismo
16.
PLoS One ; 18(9): e0292157, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37756356

RESUMO

Heparan sulfate (HS), an abundant component of the apical cell surface and basement membrane, belongs to the glycosaminoglycan family of carbohydrates covalently linked to proteins called heparan sulfate proteoglycans. After endocytosis, HS is degraded in the lysosome by several enzymes, including heparan-alpha-glucosaminide N-acetyltransferase (HGSNAT), and in its absence causes Mucopolysaccharidosis III type C (Sanfilippo type C). Since endocytosis occurs in epithelial cells of the testis and epididymis, we examined the morphological effects of Hgsnat inactivation in these organs. In the testis, Hgsnat knockout (Hgsnat-Geo) mice revealed statistically significant decrease in tubule and epithelial profile area of seminiferous tubules. Electron microscopy (EM) analysis revealed cross-sectional tubule profiles with normal and moderately to severely altered appearances. Abnormalities in Sertoli cells and blood-testis barrier and the absence of germ cells in some tubules were noted along with altered morphology of sperm, sperm motility parameters and a reduction in fertilization rates in vitro. Along with quantitatively increased epithelial and tubular profile areas in the epididymis, EM demonstrated significant accumulations of electrolucent lysosomes in the caput-cauda regions that were reactive for cathepsin D and prosaposin antibodies. Lysosomes with similar storage materials were also found in basal, clear and myoid cells. In the mid/basal region of the epithelium of caput-cauda regions of KO mice, large vacuolated cells, unreactive for cytokeratin 5, a basal cell marker, were identified morphologically as epididymal mononuclear phagocytes (eMPs). The cytoplasm of the eMPs was occupied by a gigantic lysosome suggesting an active role of these cells in removing debris from the epithelium. Some eMPs were found in proximity to T-lymphocytes, a feature of dendritic cells. Taken together, our results reveal that upon Hgsnat inactivation, morphological alterations occur to the testis affecting sperm morphology and motility parameters and abnormal lysosomes in epididymal epithelial cells, indicative of a lysosomal storage disease.

17.
J Biol Chem ; 286(33): 29417-29427, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21712390

RESUMO

Ornithine decarboxylase antizyme 3 (Oaz3) is expressed in spermatids, makes up the antizyme family of Oaz genes with Oaz1 and Oaz2, and was proposed to encode a 22 kDa antizyme protein involved in polyamine regulation similar to the 22 kDa OAZ1 and OAZ2 proteins. Here we demonstrate however that the major product encoded by Oaz3 is a 12 kDa protein, p12, which lacks the antizyme domain that interacts with ornithine decarboxylase. We show that p12 does not affect ornithine decarboxylase levels, providing an explanation for the surprising observation made in Oaz3 knock-out male mice, which do not display altered testis polyamine metabolism. This suggested a novel activity for Oaz3 p12. Using immuno-electron microscopy we localized p12 to two structures in the mammalian sperm tail, viz. the outer dense fibers and fibrous sheath, as well as to the connecting piece linking head and tail. We identified myosin phosphatase targeting subunit 3 (MYPT3), a regulator of protein phosphatase PP1ß, as a major p12-interacting protein, and show that MYPT3 is present in sperm tails and that its ankyrin repeat binds p12. We show that MYPT3 can also bind protein phosphatase PP1γ2, the only protein phosphatase present in sperm tails, and that p12- MYPT3 interaction modulates the activity of both PP1ß and PP1γ2. This is, to our knowledge, the first demonstration of a novel activity for an Oaz-encoded protein.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Proteína Fosfatase 1/metabolismo , Cauda do Espermatozoide/metabolismo , Testículo/metabolismo , Animais , Poliaminas Biogênicas/biossíntese , Proteínas de Transporte/genética , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Células NIH 3T3 , Ornitina Descarboxilase/genética , Ornitina Descarboxilase/metabolismo , Inibidores da Ornitina Descarboxilase , Proteína Fosfatase 1/genética , Estrutura Terciária de Proteína , Ratos
18.
Biol Reprod ; 86(6): 181, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22441796

RESUMO

During their epididymal maturation, stabilizing factors such as cholesterol sulfate are associated with the sperm plasma membrane. Cholesterol is sulfated in epididymal spermatozoa by the enzyme estrogen sulfotransferase. Because of its role in the efflux of sulfate conjugates formed intracellularly by sulfotransferases, the ATP-binding cassette membrane transporter G2 (ABCG2) might have a role in the translocation of this compound across the plasma membrane. In the present study we showed that ABCG2 is present in the plasma membrane overlaying the acrosomal region of spermatozoa recovered from testis, epididymis, and after ejaculation. Although ABCG2 is also present in epididymosomes, the transporter is not transferred to spermatozoa via this mechanism. Furthermore, although epididymal sperm ABCG2 was shown to be functional, as determined by its ability to extrude Hoechst 33342 in the presence of the specific inhibitor Fumitremorgin C, ABCG2 present in ejaculated sperm was found to be nonfunctional. Additional experiments demonstrated that phosphorylation of ABCG2 tyrosyl residues, but not its localization in lipid rafts, is the mechanism responsible for its functionality. Dephosphorylation of ABCG2 in ejaculated spermatozoa is proposed to cause a partial protein relocalization to other intracellular compartments. Prostasomes are proposed to have a role in this process because incubation with this fraction of seminal plasma induces a decrease in the amount of ABCG2 in the associated sperm membrane fraction. These results demonstrate that ABCG2 plays a role in epididymal sperm maturation, but not after ejaculation. The loss of ABCG2 function after ejaculation is proposed to be regulated by prostasomes.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Ejaculação , Epididimo/metabolismo , Maturação do Esperma , Espermatozoides/metabolismo , Animais , Bovinos , Masculino , Fosforilação
19.
Biol Reprod ; 86(3): 84, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22156475

RESUMO

This study arose from our finding that SubH2Bv, a histone H2B variant residing in the subacrosomal compartment of mammalian spermatozoa, contains a bipartite nuclear localization signal (bNLS) but in spite of this did not enter the spermatid nucleus. Instead, it associated with proacrosomic and acrosomic vesicles, which were targeted to the nuclear surface to form the acrosome. On this basis we proposed that SubH2Bv targets proacrosomic/acrosomic vesicles from the Golgi apparatus to the nuclear envelope by utilizing the classical bipartite/karyopherin alpha (KPNA) nuclear import pathway. To test the protein's nuclear targeting ability, SubH2Bv, with and without targeted mutations of the basic residues of bNLS, as well as bNLS alone, were transfected into mammalian cells as GFP-fusion proteins. Only the intact bNLS conferred nuclear entry. Subsequently, we showed that a KPNA, most likely KPNA6, occupies the same sperm head compartment and follows the same pattern of acrosomal association during spermiogenesis as SubH2Bv. Sperm head fractionation combined with Western blotting located this KPNA to the subacrosomal layer of the perinuclear theca, while immunocytochemistry of testicular sections showed that it associates with the surface of proacrosomic/acrosomic vesicles during acrosomal biogenesis. The identical sperm-localization and testicular-expression patterns between KPNA and SubH2Bv suggested a potential binding interaction between these proteins. This was supported by recombinant SubH2Bv affinity pull-down assays on germ cell extracts. The results of this study provide a compelling argument that these two nuclear homing proteins work in concert to direct the acrosomic vesicle to the nucleus. Their final residence in the subacrosomal layer of the perinuclear theca of spermatozoa indicates a role for SubH2Bv and KPNA in acrosomal-nuclear docking.


Assuntos
Acrossomo/fisiologia , Transporte Ativo do Núcleo Celular/fisiologia , Transdução de Sinais/fisiologia , Espermatogênese/fisiologia , alfa Carioferinas/fisiologia , Animais , Bovinos , Proteínas de Fluorescência Verde/genética , Histonas/genética , Histonas/fisiologia , Masculino , Camundongos , Mutação/genética , Cabeça do Espermatozoide/fisiologia , Transfecção
20.
Cell Tissue Res ; 349(3): 733-48, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22592626

RESUMO

As a consequence of the acrosomal reaction during fertilization, the inner acrosomal membrane (IAM) becomes exposed and forms the leading edge of the sperm for adhesive binding to and subsequent penetration of the zona-pellucida (ZP) of the metaphase-II-arrested oocyte. A premise of this review is that the IAM of spermatozoa anchors receptors and enzymes (on its extracellular side) that are required for sperm attachment to and penetration of the ZP. We propose a sperm cell fractionation strategy that allows for direct access to proteins bound to the extracellular side of the IAM. We review the types of integral and peripheral IAM proteins that have been found by this approach and that have been implicated in ZP recognition and lysis. We also propose a scheme for the origin and assembly of these proteins within the developing acrosome during spermiogenesis. During development, the extravesicular side of the membrane of the acrosomic vesicle is coated by peripheral proteins that transport and bind this secretory vesicle to the spermatid nucleus. The part of the membrane that binds to the nucleus becomes the IAM, while its extravesicular protein coat, which is retained between the IAM and the nuclear envelope of spermatozoa becomes the subacrosomal layer of the perinuclear theca (SAL-PT). Another premise of this review is that the IAM of spermatozoa is bound with proteins (on its intracellular side), namely the SAL-PT proteins, which hold the clue to the mechanism of acrosomal-nuclear docking. We propose a sperm cell fractionation strategy that allows for direct access to SAL-PT proteins. We then review the types of SAL-PT proteins that have been found by this approach and that have been implicated in transporting and binding the acrosome to the sperm nucleus.


Assuntos
Reação Acrossômica/fisiologia , Espermatogênese/fisiologia , Espermatozoides/fisiologia , Animais , Fertilização/fisiologia , Humanos , Masculino , Espermátides/metabolismo , Espermátides/fisiologia , Espermatozoides/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa