Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Mol Hum Reprod ; 29(6)2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37104740

RESUMEN

Exposure of human sperm to progesterone (P4) activates cation channel of sperm (CatSper) channels, inducing an intracellular Ca2+ concentration ([Ca2+]i) transient followed by repetitive [Ca2+]i activity (oscillations), which are believed to be functionally important. We investigated the potential significance of store-operated Ca2+-entry in these oscillations using the inhibitor SKF96365 (30 µM; SKF). Following pre-treatment of human sperm with 3 µM P4, exposure to SKF doubled the proportion of oscillating cells (P = 0.00004). In non-pre-treated cells, SKF had an effect similar to P4, inducing a [Ca2+]i transient in >80% of cells which was followed by oscillations in ≈50% of cells. The CatSper blocker RU1968 (11 µM) inhibited the SKF-induced [Ca2+]i increase and reversibly arrested [Ca2+]i oscillations. Using whole-cell patch clamp, we observed that SKF enhanced CatSper currents by 100% within 30 s, but amplitude then decayed to levels below control over the next minute. When cells were stimulated with P4, CatSper currents were stably increased (by 200%). Application of SKF then returned current amplitude to control level or less. When sperm were prepared in medium lacking bovine serum albumin (BSA), both P4 and SKF induced a [Ca2+]i transient in >95% of cells but the ability of SKF to induce oscillations was greatly reduced (P = 0.0009). We conclude that SKF, similar to a range of small organic molecules, activates CatSper channels, but that a secondary blocking action also occurs, which was detected only during patch-clamp recording. The failure of SKF to induce oscillations when cells were prepared without BSA emphasizes that the drug does not fully mimic the actions of P4.


Asunto(s)
Canales de Calcio , Señalización del Calcio , Humanos , Masculino , Canales de Calcio/metabolismo , Calcio/metabolismo , Semen/metabolismo , Motilidad Espermática , Espermatozoides/metabolismo
2.
Mol Hum Reprod ; 21(7): 563-70, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25882543

RESUMEN

Previous work has provided evidence for involvement of store-operated channels (SOCs) in [Ca(2+)]i signalling of human sperm, including a contribution to the transient [Ca(2+)]i elevation that occurs upon activation of CatSper, a sperm-specific cation channel localized to the flagellum, by progesterone. To further investigate the potential involvement of SOCs in the generation of [Ca(2+)]i signals in human sperm, we have used cell-penetrating peptides containing the important basic sequence KIKKK, part of the STIM-Orai activating region/CRAC activating domain (SOAR/CAD) of the regulatory protein stromal interaction molecule 1. SOAR/CAD plays a key role in controlling the opening of SOCs, which occurs upon mobilization of stored Ca(2+). Resting [Ca(2+)]i temporarily decreased upon application of KIKKK peptide (3-4 min), but scrambled KIKKK peptide had a similar effect, indicating that this action was not sequence-specific. However, in cells pretreated with KIKKK, the transient [Ca(2+)]i elevation induced by stimulation with progesterone decayed significantly more slowly than in parallel controls and in cells pretreated with scrambled KIKKK peptide. Examination of single-cell responses showed that this effect was due, at least in part, to an increase in the proportion of cells in which the initial transient was maintained for an extended period, lasting up to 10 min in a subpopulation of cells. We hypothesize that SOCs contribute to the progesterone-induced [Ca(2+)]i transient, and that interference with the regulatory mechanisms of SOC delays their closure, causing a prolongation of the [Ca(2+)]i transient.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Péptidos de Penetración Celular/farmacología , Progesterona/farmacología , Espermatozoides/efectos de los fármacos , Humanos , Masculino , Espermatozoides/metabolismo
3.
Hum Reprod ; 29(10): 2123-35, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25124668

RESUMEN

STUDY QUESTION: Can we identify compound(s) with reported phosphodiesterase inhibitor (PDEI) activity that could be added to human spermatozoa in vitro to enhance their motility without compromising other sperm functions? SUMMARY ANSWER: We have identified several compounds that produce robust and effective stimulation of sperm motility and, importantly, have a positive response on patient samples. WHAT IS KNOWN ALREADY: For >20 years, the use of non-selective PDEIs, such as pentoxifylline, has been known to influence the motility of human spermatozoa; however, conflicting results have been obtained. It is now clear that human sperm express several different phosphodiesterases and these are compartmentalized at different regions of the cells. By using type-specific PDEIs, differential modulation of sperm motility may be achieved without adversely affecting other functions such as the acrosome reaction (AR). STUDY DESIGN, SIZE, DURATION: This was a basic medical research study examining sperm samples from normozoospermic donors and subfertile patients attending the Assisted Conception Unit (ACU), Ninewells Hospital Dundee for diagnostic semen analysis, IVF and ICSI. Phase 1 screened 43 commercially available compounds with reported PDEI activity to identify lead compounds that stimulate sperm motility. Samples were exposed (20 min) to three concentrations (1, 10 and 100 µM) of compound, and selected candidates (n = 6) progressed to Phase 2, which provided a more comprehensive assessment using a battery of in vitro sperm function tests. PARTICIPANTS/MATERIALS, SETTING, METHODS: All healthy donors and subfertile patients were recruited at the Medical Research Institute, University of Dundee and ACU, Ninewells Hospital Dundee (ethical approval 08/S1402/6). In Phase 1, poor motility cells recovered from the 40% interface of the discontinuous density gradient were used as surrogates for patient samples. Pooled samples from three to four different donors were utilized in order to reduce variability and increase the number of cells available for simultaneous examination of multiple compounds. During Phase 2 testing, semen samples from 23 patients attending for either routine diagnostic andrology assessment or IVF/ICSI were prepared and exposed to selected compounds. Additionally, 48 aliquots of prepared samples, surplus to clinical use, were examined from IVF (n = 32) and ICSI (n = 16) patients to further determine the effects of selected compounds under clinical conditions of treatment. Effects of compounds on sperm motility were assessed by computer-assisted sperm analysis. A modified Kremer test using methyl cellulose was used to assess sperm functional ability to penetrate into viscous media. Sperm acrosome integrity and induction of apoptosis were assessed using the acrosomal content marker PSA-FITC and annexin V kit, respectively. MAIN RESULTS AND THE ROLE OF CHANCE: In Phase 1, six compounds were found to have a strong effect on poor motility samples with a magnitude of response of ≥ 60% increase in percentage total motility. Under capacitating and non-capacitating conditions, these compounds significantly (P ≤ 0.05) increased the percentage of total and progressive motility. Furthermore, these compounds enhanced penetration into a cervical mucus substitute (P ≤ 0.05). Finally, the AR was not significantly induced and these compounds did not significantly increase the externalization of phosphatidylserine (P = 0.6, respectively). In general, the six compounds maintained the stimulation of motility over long periods of time (180 min) and their effects were still observed after their removal. In examinations of clinical samples, there was a general observation of a more significant stimulation of sperm motility in samples with lower baseline motility. In ICSI samples, compounds #26, #37 and #38 were the most effective at significantly increasing total motility (88, 81 and 79% of samples, respectively) and progressive motility (94, 93 and 81% of samples, respectively). In conclusion, using a two-phased drug discovery screening approach including the examination of clinical samples, 3/43 compounds were identified as promising candidates for further study. LIMITATIONS, REASONS FOR CAUTION: This is an in vitro study and caution must be taken when extrapolating the results. Data for patients were from one assessment and thus the robustness of responses needs to be established. The n values for ICSI samples were relatively small. WIDER IMPLICATIONS OF THE FINDINGS: We have systematically screened and identified several compounds that have robust and effective stimulation (i.e. functional significance with longevity and no toxicity) of total and progressive motility under clinical conditions of treatment. These compounds could be clinical candidates with possibilities in terms of assisted reproductive technology options for current or future patients affected by asthenozoospermia or oligoasthenozoospermia. STUDY FUNDING/COMPETING INTERESTS: This study was funded primarily by the MRC (DPFS) but with additional funding from the Wellcome Trust, Tenovus (Scotland), University of Dundee, NHS Tayside and Scottish Enterprise. The authors have no competing interests. A patent (#WO2013054111A1) has been published containing some of the information presented in this manuscript.


Asunto(s)
Inhibidores de Fosfodiesterasa/farmacología , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Acrosoma/efectos de los fármacos , Apoptosis/efectos de los fármacos , Humanos , Masculino , Espermatozoides/fisiología
4.
Biochem J ; 448(2): 189-200, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22943284

RESUMEN

Ca2+i signalling is pivotal to sperm function. Progesterone, the best-characterized agonist of human sperm Ca2+i signalling, stimulates a biphasic [Ca2+]i rise, comprising a transient and subsequent sustained phase. In accordance with recent reports that progesterone directly activates CatSper, the [Ca2+]i transient was detectable in the anterior flagellum (where CatSper is expressed) 1-2 s before responses in the head and neck. Pre-treatment with 5 µM 2-APB (2-aminoethoxydiphenyl borate), which enhances activity of store-operated channel proteins (Orai) by facilitating interaction with their activator [STIM (stromal interaction molecule)] 'amplified' progesterone-induced [Ca2+]i transients at the sperm neck/midpiece without modifying kinetics. The flagellar [Ca2+]i response was unchanged. 2-APB (5 µM) also enhanced the sustained response in the midpiece, possibly reflecting mitochondrial Ca2+ accumulation downstream of the potentiated [Ca2+]i transient. Pre-treatment with 50-100 µM 2-APB failed to potentiate the transient and suppressed sustained [Ca2+]i elevation. When applied during the [Ca2+]i plateau, 50-100 µM 2-APB caused a transient fall in [Ca2+]i, which then recovered despite the continued presence of 2-APB. Loperamide (a chemically different store-operated channel agonist) enhanced the progesterone-induced [Ca2+]i signal and potentiated progesterone-induced hyperactivated motility. Neither 2-APB nor loperamide raised pHi (which would activate CatSper) and both compounds inhibited CatSper currents. STIM and Orai were detected and localized primarily to the neck/midpiece and acrosome where Ca2+ stores are present and the effects of 2-APB are focussed, but store-operated currents could not be detected in human sperm. We propose that 2-APB-sensitive channels amplify [Ca2+]i elevation induced by progesterone (and other CatSper agonists), amplifying, propagating and providing spatio-temporal complexity in [Ca2+]i signals of human sperm.


Asunto(s)
Compuestos de Boro/farmacología , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Agonistas de los Canales de Calcio/farmacología , Moléculas de Adhesión Celular/metabolismo , Humanos , Técnicas In Vitro , Loperamida/farmacología , Masculino , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1 , Proteína ORAI2 , Progesterona/farmacología , Pieza Intermedia del Espermatozoide/efectos de los fármacos , Pieza Intermedia del Espermatozoide/metabolismo , Motilidad Espermática/efectos de los fármacos , Molécula de Interacción Estromal 1 , Molécula de Interacción Estromal 2
5.
Reprod Biol ; 21(4): 100561, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34619633

RESUMEN

Human spermatozoa interact with a complex biochemical environment in the female reproductive tract en route to the site of fertilisation. Ovarian follicular fluid contributes to this complex milieu and is known to contain steroids such as progesterone, whose effects on sperm physiology have been widely characterised. We have previously reported that progesterone stimulates intracellular calcium concentration ([Ca2+]i) signalling and acrosome reaction in human spermatozoa. To characterise the effects of the unified complete follicular fluid steroid hormone complement on human spermatozoa, a comprehensive, data-based, 'physiological standard' steroid hormone balance of follicular fluid (shFF) was created from individual constituents. shFF induced a rapid biphasic [Ca2+]i elevation in human spermatozoa. Using population fluorimetry, we compared [Ca2+]i signal amplitude in cells exposed to serial applications of shFF (6 steps from 10-5X up to 1X shFF) with responses to the equivalent progesterone component alone (6 steps from 135 pM - 13.5µM). Threshold for the response to shFF was right-shifted (≈10-fold) compared to progesterone alone, but the maximum response to shFF was greatly enhanced. An acrosome reaction assay was used to assess functional effects of shFF-induced sperm calcium signalling. shFF as well as progesterone-treated spermatozoa showed a significant increase in % acrosome reaction (P < 0.01). All of this evidence suggests the modulation of progesterone-mediated responses by other follicular fluid steroids.


Asunto(s)
Líquido Folicular/química , Hormonas/fisiología , Espermatozoides/fisiología , Reacción Acrosómica , Calcio/metabolismo , Femenino , Hormonas/análisis , Humanos , Masculino
6.
Reproduction ; 138(3): 425-37, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19542252

RESUMEN

Intracellular Ca2+ stores play a central role in the regulation of cellular [Ca2+](i) and the generation of complex [Ca2+] signals such as oscillations and waves. Ca2+ signalling is of particular significance in sperm cells, where it is a central regulator in many key activities (including capacitation, hyperactivation, chemotaxis and acrosome reaction) yet mature sperm lack endoplasmic reticulum and several other organelles that serve as Ca2+ stores in somatic cells. Here, we review i) the evidence for the expression in sperm of the molecular components (pumps and channels) which are functionally significant in the activity of Ca2+ stores of somatic cells and ii) the evidence for the existence of functional Ca2+ stores in sperm. This evidence supports the existence of at least two storage organelles in mammalian sperm, one in the acrosomal region and another in the region of the sperm neck and midpiece. We then go on to discuss the probable identity of these organelles and their discrete functions: regulation by the acrosome of its own secretion and regulation by membranous organelles at the sperm neck (and possibly by the mitochondria) of flagellar activity and hyperactivation. Finally, we consider the ability of the sperm discretely to control mobilisation of these stores and the functional interaction of stored Ca2+ at the sperm neck/midpiece with CatSper channels in the principal piece in regulation of the activities of mammalian sperm.


Asunto(s)
Calcio/metabolismo , Calcio/fisiología , Espermatozoides/metabolismo , Espermatozoides/fisiología , Animales , Canales de Calcio/metabolismo , Canales de Calcio/fisiología , Señalización del Calcio/fisiología , Humanos , Masculino , Modelos Biológicos , Orgánulos/metabolismo , Orgánulos/fisiología , Espermatozoides/citología
7.
Int J Dev Biol ; 52(5-6): 615-26, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18649275

RESUMEN

Calcium signalling plays a pivotal role in sperm physiology, being intimately involved in the regulation of acrosome reaction, chemotaxis and hyperactivation. Here we describe briefly the mechanisms of calcium regulation in somatic cells and the ways in which these mechanisms have been adapted to function in mature spermatozoa. We then consider recent data from this and other laboratories on the responses of sperm to three compounds: progesterone and nitric oxide (both products of the cumulus oophorus) and 4-aminopyridine. All of these compounds induce calcium signals in the posterior sperm head and neck region and, when applied at appropriate concentrations, modify flagellar activity, causing asymmetric bending of the proximal flagellum. We argue that these effects reflect a common mode of action, mobilisation of calcium stored in the sperm neck region. Finally we consider the nature of calcium signalling pathways in sperm. We suggest that this highly specialised and extremely polarised cell, though working with the same calcium signalling 'tools' as those of somatic cells, employs them to generate unusually 'hard-wired' calcium signals that do not act to integrate stimuli. 'Leakage' between these calcium signalling pathways will generate inappropriate responses, compromising functioning of the cell.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Cola del Espermatozoide/metabolismo , Espermatozoides/fisiología , 4-Aminopiridina/química , 4-Aminopiridina/metabolismo , Humanos , Masculino , Modelos Biológicos , Óxido Nítrico/metabolismo , Progesterona/metabolismo , Transducción de Señal , Motilidad Espermática , Espermatozoides/metabolismo
9.
Theriogenology ; 63(2): 599-614, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15626419

RESUMEN

It is generally accepted that sperm capacitation is associated with the protein kinase A-mediated appearance of tyrosine phosphoproteins, although the substrates and kinase(s) involved have not been identified. We described a Mr 32,000 tyrosine phosphoprotein, "p32", appearing in porcine sperm coincident with capacitation. We also discovered a tyrosine kinase-like enzyme in boar sperm of Mr 32,000 ("TK-32") with enhanced activity during capacitation. The present work was conducted to further characterize and to identify these capacitation-related protein(s). Fresh porcine sperm were incubated to induce capacitation then immunoprecipitation, immunoblotting and proteomic analysis revealed seven tyrosine-phosphorylated proteins aligned in the range of Mr 30,000 with different isoelectric pH values (pI). Therefore, p32 may be composed of several tyrosine phosphoproteins. Three were identified as acrosin-binding sp32 (pI 6.5), and two triosephosphate isomerase isoforms (pI 7.1 and 7.9). At present, however, proteonomic analysis has not revealed any kinase at Mr 32,000. Immunoprecipitation experiments show that p32 and TK-32 are different molecules, as TK-32 activity remains in the supernatant of the antiphosphotyrosine precipitates. Finally, in-gel renaturation and immunoblotting suggest that TK-32 is a mitogen-activated protein kinase (MAPK). The discovery of p32 and the MAPK-like TK-32 provides new insight regarding the mechanisms underlying capacitation in the pig.


Asunto(s)
Fosfoproteínas , Proteínas Tirosina Quinasas/metabolismo , Proteómica/métodos , Capacitación Espermática/fisiología , Porcinos , Animales , Electroforesis en Gel Bidimensional , Masculino , Peso Molecular , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosfotirosina/química , Fosfotirosina/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
10.
J Androl ; 23(5): 709-16, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12185106

RESUMEN

Spermatozoa undergo a variety of changes during their life that are prerequisites to their maturation and ability to fertilize eggs. Mammalian sperm capacitation and acrosome reaction are regulated by signal transduction systems involving cyclic adenosine monophosphate (cAMP) as a second messenger. This second messenger acts through the activation of protein kinase A (PKA) and indirectly regulates protein tyrosine phosphorylation. cAMP levels are controlled by a balance of phosphodiesterases (PDEs) and adenylyl cyclase (AC) enzymatic activities, which are responsible for its degradation and production, respectively. The aim of this study was to evaluate the possible relationship between the intracellular levels of cAMP and PDE and PKA activities during human sperm capacitation induced by fetal cord serum ultrafiltrate (FCSu) and acrosome reaction induced by calcium ionophore A23187. We report that PKA activity was higher in capacitating than in noncapacitating spermatozoa and that intracellular levels of cAMP decreased but that PDE activity remained constant during capacitation. The acrosome reaction induced by A23187 was associated with increases in cAMP and PKA activity but not in PDE activity. These results strongly suggest that net cAMP concentration is under the control of AC, since PDE activity is constant during sperm capacitation and the acrosome reaction. Moreover, the results suggest that low levels of cAMP are sufficient for capacitation and PKA activation and/or that the cAMP concentration measured in whole spermatozoa does not reflect the effective intracellular cAMP levels present in specific compartments of these cells.


Asunto(s)
Reacción Acrosómica/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Capacitación Espermática/fisiología , Calcimicina/farmacología , AMP Cíclico/metabolismo , Activación Enzimática , Humanos , Membranas Intracelulares/metabolismo , Ionóforos/farmacología , Masculino , Hidrolasas Diéster Fosfóricas/metabolismo , Espermatozoides/efectos de los fármacos , Espermatozoides/fisiología , Fracciones Subcelulares/metabolismo , Distribución Tisular
11.
J Vis Exp ; (40)2010 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-20567212

RESUMEN

Fluorescence microscopy of cells loaded with fluorescent, Ca(2+)-sensitive dyes is used for measurement of spatial and temporal aspects of Ca(2+) signaling in live cells. Here we describe the method used in our laboratories for loading suspensions of human sperm with Ca(2+)-reporting dyes and measuring the fluorescence signal during physiological stimulation. Motile cells are isolated by direct swim-up and incubated under capacitating conditions for 0-24 h, depending upon the experiment. The cell-permeant AM (acetoxy methyl ester) ester form of the Ca(2+)-reporting dye is then added to a cell aliquot and a period of 1 h is allowed for loading of the dye into the cytoplasm. We use visible wavelength dyes to minimize photo-damage to the cells, but this means that ratiometric recording is not possible. Advantages and disadvantages of this approach are discussed. During the loading period cells are introduced into an imaging chamber and allowed to adhere to a poly-D-lysine coated coverslip. At the end of the loading period excess dye and loose cells are removed by connection of the chamber to the perfusion apparatus. The chamber is perfused continuously, stimuli and modified salines are then added to the perfusion header. Experiments are recorded by time-lapse acquisition of fluorescence images and analyzed in detail offline, by manually drawing regions of interest. Data are normalized to pre-stimulus levels such that, for each cell (or part of a cell), a graph showing the Ca(2+) response as % change in fluorescence is obtained.


Asunto(s)
Señalización del Calcio , Calcio/análisis , Microscopía Fluorescente/métodos , Espermatozoides/metabolismo , Calcio/metabolismo , Humanos , Masculino , Espermatozoides/química
12.
Commun Integr Biol ; 2(2): 82-5, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19704895

RESUMEN

Signaling through [Ca(2+)](i) is central to regulation of sperm activity and is likely to be the mechanism that transduces signals from the female reproductive tract to regulate sperm motility. In a recent paper1 we showed that exposure of sperm to nitric oxide mobilizes stored Ca(2+) in human sperm, an effect that occurs through nitrosylation of protein thiols. Not only did we find that NO* production by cells of the human female tract would be sufficient to elicit this effect, but progesterone, which is also present in the female tract and is synthesized by the oocyte vestments, acted synergistically with NO* to mobilize Ca(2+) and enhance flagellar beating. Here we argue that a Ca(2+) store at the junction of the sperm head and flagellum is subject to regulation by both progesterone and NO* and that ryanodine receptors at the store may be the point at which coincidence detection and synergistic interaction occurs.

13.
Development ; 135(22): 3677-86, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18842814

RESUMEN

Generation of NO by nitric oxide synthase (NOS) is implicated in gamete interaction and fertilisation. Exposure of human spermatozoa to NO donors caused mobilisation of stored Ca(2+) by a mechanism that did not require activation of guanylate cyclase but was mimicked by S-nitroso-glutathione (GSNO; an S-nitrosylating agent). Application of dithiothreitol, to reduce protein -SNO groups, rapidly reversed the actions of NO and GSNO on [Ca(2+)](i). The effects of NO, GSNO and dithiothreitol on sperm protein S-nitrosylation, assessed using the biotin switch method, closely paralleled their actions on [Ca(2+)](i). Immunofluorescent staining revealed constitutive and inducible NOS in human oviduct and cumulus (the cellular layer investing the oocyte). 4,5-diaminofluorescein (DAF) staining demonstrated production of NO by these tissues. Incubation of human sperm with oviduct explants induced sperm protein S-nitrosylation resembling that induced by NO donors and GSNO. Progesterone (a product of cumulus cells) also mobilises stored Ca(2+) in human sperm. Pre-treatment of sperm with NO greatly enhanced the effect of progesterone on [Ca(2+)](i), resulting in a prolonged increase in flagellar excursion. We conclude that NO regulates mobilisation of stored Ca(2+) in human sperm by protein S-nitrosylation, that this action is synergistic with that of progesterone and that this synergism is potentially highly significant in gamete interactions leading to fertilisation.


Asunto(s)
Calcio/metabolismo , Trompas Uterinas/metabolismo , Flagelos/metabolismo , Óxido Nítrico/biosíntesis , Espermatozoides/metabolismo , Células del Cúmulo/enzimología , Ditiotreitol/farmacología , Femenino , Glutatión/metabolismo , Guanilato Ciclasa/metabolismo , Humanos , Cinética , Masculino , Óxido Nítrico Sintasa/metabolismo , Progesterona/farmacología , Espermatozoides/efectos de los fármacos
15.
Proteomics ; 7(17): 3066-84, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17683036

RESUMEN

Nitric oxide (NO) enhances human sperm motility and capacitation associated with increased protein phosphorylation. NO activates soluble guanylyl cyclase, but can also modify protein function covalently via S-nitrosylation of cysteine. Remarkably, this mechanism remains unexplored in sperm although they depend on post-translational protein modification to achieve changes in function required for fertilisation. Our objective was to identify targets for S-nitrosylation in human sperm. Spermatozoa were incubated with NO donors and S-nitrosylated proteins were identified using the biotin switch assay and a proteomic approach using MS/MS. 240 S-nitrosylated proteins were detected in sperm incubated with S-nitroso-glutathione. Minimal levels were observed in glutathione or untreated samples. Proteins identified consistently based on multiple peptides included established targets for S-nitrosylation in other cells e.g. tubulin, GST and HSPs but also novel targets including A-kinase anchoring protein (AKAP) types 3 and 4, voltage-dependent anion-selective channel protein 3 and semenogelin 1 and 2. In situ localisation revealed S-nitrosylated targets on the postacrosomal region of the head and throughout the flagellum. Potential targets for S-nitrosylation in human sperm include physiologically significant proteins not previously reported in other cells. Their identification will provide novel insight into the mechanism of action of NO in spermatozoa.


Asunto(s)
Óxido Nítrico/metabolismo , Compuestos Nitrosos/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Espermatozoides/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cisteína/metabolismo , Humanos , Masculino , Proteínas de Transporte de Membrana Mitocondrial , Donantes de Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteoma/química , S-Nitrosoglutatión/metabolismo , S-Nitrosoglutatión/farmacología , Proteínas de Secreción de la Vesícula Seminal/metabolismo , Espectrometría de Masas en Tándem , Canales Aniónicos Dependientes del Voltaje/metabolismo
16.
Reproduction ; 133(4): 675-84, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17504912

RESUMEN

Although sperm dysfunction is the single most common cause of infertility, we have poor methods of diagnosis and surprisingly no effective treatment (excluding assisted reproductive technology). In this review, we challenge the usefulness of a basic semen analysis and argue that a new paradigm is required immediately. We discuss the use of at-home screening to potentially improve the diagnosis of the male and to streamline the management of the sub-fertile couple. Additionally, we outline the recent progress in the field, for example, in proteomics, which will allow the development of new biomarkers of sperm function. This new knowledge will transform our understanding of the spermatozoon as a machine and is likely to lead to non-ART treatments for men with sperm dysfunction.


Asunto(s)
Infertilidad Masculina/diagnóstico , Espermatozoides/fisiología , Calcio/metabolismo , Humanos , Masculino , Proteómica , Capacitación Espermática/fisiología , Recuento de Espermatozoides , Espermatozoides/citología
17.
J Cell Sci ; 118(Pt 8): 1673-85, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15811949

RESUMEN

The sarcoplasmic-endoplasmic reticulum Ca(2+)-ATPase (SERCA) inhibitors thapsigargin (0.1-1 microM) and cyclopiazonic acid (10 microM), failed to affect resting [Ca(2+)] in human spermatozoa. Slow progesterone-induced [Ca(2+ i)](i) oscillations in human spermatozoa, which involve cyclic emptying-refilling of an intracellular Ca(2+) store were also insensitive to these inhibitors. Non-selective doses of thapsigargin (5-30 microM, 50-300 times the saturating dose for SERCA inhibition), caused elevation of resting [Ca(2+)](i) and partial, dose-dependent disruption of oscillations. A 10-40 microM concentration of bis(2-hydroxy-3-tert-butyl-5-methyl-phenyl)methane (bis-phenol), which inhibits both thapsigargin-sensitive and -insensitive microsomal Ca(2+) ATPases, caused elevation of resting [Ca(2+)](i) and inhibition of [Ca(2+)](i) oscillations at doses consistent with inhibition of thapsigargin-resistant, microsomal ATPase and liberation of stored Ca(2+). Low doses of bis-phenol had marked effects on [Ca(2+)](i) oscillation kinetics. Application of the drug to cells previously stimulated with progesterone had effects very similar to those observed when it was applied to unstimulated cells, suggesting that the sustained Ca(2+) influx induced by progesterone is not mediated via mobilisation of Ca(2+) stores. Western blotting for human sperm proteins showed expression of secretory pathway Ca(2+) ATPase (SPCA1). Immunolocalisation studies revealed expression of SPCA1 in all cells in an area behind the nucleus, extending into the midpiece. Staining for SERCA, carried out in parallel, detected no expression with either technique. We conclude that: (1) intracellular Ca(2+) store(s) and store-dependent [Ca(2+)](i) oscillations in human spermatozoa rely primarily on a thapsigargin/cyclopiazonic acid-insensitive Ca(2+) pump, which is not a SERCA as characterised in somatic cells; (2) effects of high-dose thapsigargin on spermatozoa primarily reflect non-specific actions on non-SERCAs and; (3) secretory pathway Ca(2+) ATPases contribute at least part of this non-SERCA Ca(2+) pump activity.


Asunto(s)
Señalización del Calcio/fisiología , ATPasas Transportadoras de Calcio/metabolismo , Calcio/metabolismo , Espermatozoides/enzimología , Hidroxitolueno Butilado/análogos & derivados , Hidroxitolueno Butilado/farmacología , Señalización del Calcio/efectos de los fármacos , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , Compartimento Celular/efectos de los fármacos , Compartimento Celular/fisiología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Citoplasma/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Inmunohistoquímica , Líquido Intracelular/metabolismo , Masculino , Manganeso/metabolismo , Progesterona/farmacología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Tapsigargina/farmacología
18.
Mol Reprod Dev ; 69(4): 428-35, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15457543

RESUMEN

Second messengers are involved in sperm fertilizing potential, as both motility and the acrosome reaction are influenced by cAMP. Moreover, the activity of cyclic nucleotides is implicated in the appearance of tyrosine phosphorylated sperm proteins, which is associated with capacitation in the mammalian spermatozoa. Nevertheless, the involvement of the cAMP/protein kinase A (PK-A) pathway during pig sperm capacitation may be different from that observed in other mammals. The objective of the present study was to clarify the cAMP/PK-A pathway during the capacitation of porcine spermatozoa and to evaluate this impact on the p32 sperm tyrosine phosphoprotein appearance. The presence of p32 was assessed after incubating fresh pig sperm with IBMX/db-cAMP, H-89, a PK-A inhibitor or bistyrphostin, a tyrosine kinase inhibitor, in capacitating (CM) or non-capacitating conditions (NCM) by immunoblotting SDS-extracted and separated sperm proteins using an anti-phosphotyrosine antibody. When pig spermatozoa were incubated in CM supplemented with H-89 (50 microM) or bistyrphostin (1.2 microM), capacitation decreased significantly (P < 0.001). The p32 sperm tyrosine phosphoprotein, previously shown to be associated with capacitation of porcine sperm though not necessarily an end point of this phenomenon, was not modulated by IBMX/db-cAMP (100 microM/1 mM), H-89 (50 microM) nor bistyrphostin (1.2 microM). Our results indicate, therefore, that pig sperm are regulated somewhat differently than as described for other mammals, because although the cAMP/PK-A and tyrosine kinase pathways are involved in capacitation, they do not influence the appearance of p32.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Sistemas de Mensajero Secundario/fisiología , Capacitación Espermática/fisiología , Espermatozoides/fisiología , Animales , Células Cultivadas , Masculino , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Fosfotirosina/metabolismo , Porcinos
19.
Biol Reprod ; 67(2): 423-30, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12135876

RESUMEN

Mammalian sperm motility, capacitation, and the acrosome reaction are regulated by signal transduction systems involving cAMP as a second messenger. Levels of cAMP are controlled by two key enzymes, adenylyl cyclase and phosphodiesterases (PDEs), the latter being involved in cAMP degradation. Calmodulin-dependent PDE (PDE1) and cAMP-specific PDE (PDE4) activities were previously identified in spermatozoa via the use of specific inhibitors. Here we report that human sperm PDEs are associated with the plasma membrane (50%-60%) as well as with the particulate fraction (30%-50%) and have more affinity for cAMP than cGMP. Immunocytochemical data indicated that PDE1A, a variant of PDE1, is localized on the equatorial segment of the sperm head as well as on the mid and principal pieces of the flagellum, and that PDE3A is found on the postacrosomal segment of the sperm head. Immunoblotting confirmed the presence of PDE1A and PDE3A isoforms in spermatozoa. Milrinone, a PDE3 inhibitor, increased intracellular levels of cAMP by about 15% but did not affect sperm functions, possibly because PDE3 represents only a small proportion of the sperm total PDE activity (10% and 25% in Triton X-100 soluble and particulate fractions, respectively). PDE1A activity in whole sperm extract or after partial purification by anion-exchange chromatography was not stimulated by calcium + calmodulin. Results obtained with electrophoresis in native conditions indicated that calmodulin is tightly bound to PDE1A. Incubation with EGTA + EDTA, trifluoperazine, or urea did not dissociate the PDE1A-calmodulin complex. These results suggest that PDE1A is permanently activated in human spermatozoa.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Calmodulina/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Espermatozoides/enzimología , 1-Metil-3-Isobutilxantina/farmacología , Adulto , Cromatografía por Intercambio Iónico , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1 , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Electroforesis en Gel de Poliacrilamida , Humanos , Immunoblotting , Inmunohistoquímica , Técnicas In Vitro , Masculino , Milrinona/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Capacitación Espermática , Espermatozoides/metabolismo
20.
J Cell Sci ; 115(Pt 2): 311-20, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11839783

RESUMEN

Surf clam (Spisula solidissima) oocytes are spawned at the prophase I stage of meiosis, and they remain arrested at this stage until fertilization. Full oocyte meiosis reinitiation, first evidenced by germinal vesicle breakdown (GVBD), may be induced by artificial activators mimicking sperm, such as high K(+) or serotonin. Previous reports indicated that treatments thought to increase the level of oocyte cAMP inhibited sperm- or serotonin-induced, but not KCl-induced, GVBD in clam oocytes. These observations extend the well known requirement for a drop in oocyte cAMP levels in mammalian, amphibian or starfish oocytes and support the view that such a drop is universally important throughout the animal kingdom. We have re-examined the cAMP dependency of GVBD in clam oocytes and found that various treatments that raise oocyte cAMP levels did not, surprisingly, affect either KCl- or serotonin-induced GVBD. Such treatments, however, inhibited GVBD upon insemination of the oocytes, but this was due to the failure of sperm to fuse/penetrate the oocytes; thus, it was not an inhibition of oocyte activation as such. Direct measurements of oocyte cAMP levels after activation by serotonin, KCl or sperm showed that, contrary to expectations, there is a rise in cAMP levels before GVBD. Using SQ22536, an adenylyl cyclase inhibitor, the increase in oocyte cAMP level was partly prevented and GVBD proceeded, but with a significant retardation, indicating that the normal cAMP rise facilitates GVBD. Our work sheds light on the diversity of upstream pathways leading to activation of MPF and provides a unique model whereby the onset of meiosis reinitiation is associated with an increase, not a decrease, in oocyte cAMP levels.


Asunto(s)
Adenina/análogos & derivados , Bivalvos/metabolismo , AMP Cíclico/metabolismo , Vesículas Citoplasmáticas/metabolismo , Meiosis/fisiología , Oocitos/metabolismo , Profase/fisiología , Regulación hacia Arriba/genética , 1-Metil-3-Isobutilxantina/farmacología , Adenina/farmacología , Animales , Bivalvos/citología , Bivalvos/genética , Colforsina/farmacología , Vesículas Citoplasmáticas/efectos de los fármacos , Vesículas Citoplasmáticas/genética , Interacciones Farmacológicas/fisiología , Inhibidores Enzimáticos/farmacología , Femenino , Fertilización/efectos de los fármacos , Fertilización/fisiología , Meiosis/efectos de los fármacos , Oocitos/citología , Oocitos/efectos de los fármacos , Cloruro de Potasio/metabolismo , Cloruro de Potasio/farmacología , Profase/efectos de los fármacos , Serotonina/metabolismo , Serotonina/farmacología , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA