Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 149(2)2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34919125

RESUMEN

The CatSper cation channel is essential for sperm capacitation and male fertility. The multi-subunit CatSper complexes form highly organized calcium signaling nanodomains on flagellar membranes. Here, we report identification of an uncharacterized protein, C2CD6, as a subunit of the mouse CatSper complex. C2CD6 contains a calcium-dependent, membrane-targeting C2 domain. C2CD6 associates with the CatSper calcium-selective, core-forming subunits. Deficiency of C2CD6 depletes the CatSper nanodomains from the flagellum and results in male sterility. C2CD6-deficient sperm are defective in hyperactivation and fail to fertilize oocytes both in vitro and in vivo. CatSper currents are present but at a significantly lower level in C2CD6-deficient sperm. Transient treatments with either Ca2+ ionophore, starvation, or a combination of both restore the fertilization capacity of C2CD6-deficient sperm. C2CD6 interacts with EFCAB9, a pH-dependent calcium sensor in the CatSper complex. We postulate that C2CD6 facilitates incorporation of the CatSper complex into the flagellar plasma membrane and may function as a calcium sensor. The identification of C2CD6 may enable the long-sought reconstitution of the CatSper ion channel complex in a heterologous system for male contraceptive development.


Asunto(s)
Canales de Calcio , Infertilidad Masculina , Cola del Espermatozoide , Animales , Femenino , Masculino , Ratones , Potenciales de Acción , Calcio/metabolismo , Canales de Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Infertilidad Masculina/genética , Ratones Endogámicos C57BL , Multimerización de Proteína , Transporte de Proteínas , Motilidad Espermática , Cola del Espermatozoide/metabolismo , Cola del Espermatozoide/fisiología
2.
Mol Hum Reprod ; 30(3)2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38341666

RESUMEN

To become fertile, mammalian sperm are required to undergo capacitation in the female tract or in vitro in defined media containing ions (e.g. HCO3 -, Ca2+, Na+, and Cl-), energy sources (e.g. glucose, pyruvate) and serum albumin (e.g. bovine serum albumin (BSA)). These different molecules initiate sequential and concomitant signaling pathways, leading to capacitation. Physiologically, capacitation induces changes in the sperm motility pattern (e.g. hyperactivation) and prepares sperm for the acrosomal reaction (AR), two events required for fertilization. Molecularly, HCO3 - activates the atypical adenylyl cyclase Adcy10 (aka sAC), increasing cAMP and downstream cAMP-dependent pathways. BSA, on the other hand, induces sperm cholesterol release as well as other signaling pathways. How these signaling events, occurring in different sperm compartments and with different kinetics, coordinate among themselves is not well established. Regarding the AR, recent work has proposed a role for glycogen synthase kinases (GSK3α and GSK3ß). GSK3α and GSK3ß are inactivated by phosphorylation of residues Ser21 and Ser9, respectively, in their N-terminal domain. Here, we present evidence that GSK3α (but not GSK3ß) is present in the anterior head and that it is regulated during capacitation. Interestingly, BSA and HCO3 - regulate GSK3α in opposite directions. While BSA induces a fast GSK3α Ser21 phosphorylation, HCO3 - and cAMP-dependent pathways dephosphorylate this residue. We also show that the HCO3--induced Ser21 dephosphorylation is mediated by hyperpolarization of the sperm plasma membrane potential (Em) and by intracellular pH alkalinization. Previous reports indicate that GSK3 kinases mediate the progesterone-induced AR. Here, we show that GSK3 inhibition also blocks the Ca2+ ionophore ionomycin-induced AR, suggesting a role for GSK3 kinases downstream of the increase in intracellular Ca2+ needed for this exocytotic event. Altogether, our data indicate a temporal and biphasic GSK3α regulation with opposite actions of BSA and HCO3 -. Our results also suggest that this regulation is needed to orchestrate the AR during sperm capacitation.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Albúmina Sérica Bovina , Capacitación Espermática , Animales , Femenino , Masculino , Ratones , Calcio/metabolismo , AMP Cíclico/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Mamíferos , Fosforilación , Semen/metabolismo , Albúmina Sérica Bovina/farmacología , Albúmina Sérica Bovina/metabolismo , Motilidad Espermática , Espermatozoides/metabolismo
3.
Reproduction ; 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38912971

RESUMEN

Valosin-containing protein (VCP; aka p97), a member of the AAA family (ATPases Associated with various cellular Activities), has been associated with a wide range of cellular functions. While previous evidence has shown its presence in mammalian sperm, our study unveils its function in mouse sperm. Notably, we found that mouse VCP does not undergo tyrosine phosphorylation during capacitation and exhibits distinct localization patterns. In the sperm head, it resides within the equatorial segment and, following exocytosis, it is released and cleaved. In the flagellum, VCP is observed in the principal and midpiece. Furthermore, our research highlights a unique role for VCP in the cAMP/PKA pathway during capacitation. Pharmacological inhibition of sperm VCP led to reduced intracellular cAMP levels that resulted in decreased phosphorylation in PKA substrates and tyrosine residues, and diminished fertilization competence. Our results show that in mouse sperm, VCP plays a pivotal role in regulating cAMP production, probably by the modulation of soluble adenylyl cyclase (sAC) activity.

4.
Biol Reprod ; 109(3): 340-355, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37399121

RESUMEN

ADAD1 is a testis-specific RNA-binding protein expressed in post-meiotic spermatids whose loss leads to defective sperm and male infertility. However, the drivers of the Adad1 phenotype remain unclear. Morphological and functional analysis of Adad1 mutant sperm showed defective DNA compaction, abnormal head shaping, and reduced motility. Mutant testes demonstrated minimal transcriptome changes; however, ribosome association of many transcripts was reduced, suggesting ADAD1 may be required for their translational activation. Further, immunofluorescence of proteins encoded by select transcripts showed delayed protein accumulation. Additional analyses demonstrated impaired subcellular localization of multiple proteins, suggesting protein transport is also abnormal in Adad1 mutants. To clarify the mechanism giving rise to this, the manchette, a protein transport microtubule network, and the LINC (linker of nucleoskeleton and cytoskeleton) complex, which connects the manchette to the nuclear lamin, were assessed across spermatid development. Proteins of both displayed delayed translation and/or localization in mutant spermatids implicating ADAD1 in their regulation, even in the absence of altered ribosome association. Finally, ADAD1's impact on the NPC (nuclear pore complex), a regulator of both the manchette and the LINC complex, was examined. Reduced ribosome association of NPC encoding transcripts and reduced NPC protein abundance along with abnormal localization in Adad1 mutants confirmed ADAD1's impact on translation is required for a NPC in post-meiotic germ cells. Together, these studies lead to a model whereby ADAD1's influence on nuclear transport leads to deregulation of the LINC complex and the manchette, ultimately generating the range of physiological defects observed in the Adad1 phenotype.


Asunto(s)
Poro Nuclear , Espermátides , Ratones , Animales , Masculino , Espermátides/metabolismo , Poro Nuclear/genética , Poro Nuclear/metabolismo , Proteínas Portadoras/metabolismo , Semen/metabolismo , Espermatozoides/metabolismo , Espermatogénesis/genética , Testículo/metabolismo , Proteínas/metabolismo , Microtúbulos/metabolismo
5.
FASEB J ; 35(6): e21478, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33991146

RESUMEN

Sperm capacitation is essential to gain fertilizing capacity. During this process, a series of biochemical and physiological modifications occur that allow sperm to undergo acrosomal exocytosis (AE). At the molecular level, hyperpolarization of the sperm membrane potential (Em) takes place during capacitation. This study shows that human sperm incubated under conditions that do not support capacitation (NC) can become ready for an agonist stimulated AE by pharmacologically inducing Em hyperpolarization with Valinomycin or Amiloride. To investigate how Em hyperpolarization promotes human sperm's ability to undergo AE, live single-cell imaging experiments were performed to simultaneously monitor changes in [Ca2+ ]i and the occurrence of AE. Em hyperpolarization turned [Ca2+ ]i dynamics in NC sperm from spontaneously oscillating into a sustained slow [Ca2+ ]i increase. The addition of progesterone (P4) or K+ to Valinomycin-treated sperm promoted that a significant number of cells displayed a transitory rise in [Ca2+ ]i which then underwent AE. Altogether, our results demonstrate that Em hyperpolarization is necessary and sufficient to prepare human sperm for the AE. Furthermore, this Em change decreased Ca2+ oscillations that block the occurrence of AE, providing strong experimental evidence of the molecular mechanism that drives the acquisition of acrosomal responsiveness.


Asunto(s)
Reacción Acrosómica , Señalización del Calcio , Exocitosis , Potenciales de la Membrana , Capacitación Espermática , Espermatozoides/fisiología , Humanos , Masculino , Fosforilación
6.
FASEB J ; 35(4): e21528, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33742713

RESUMEN

We have recently reported two different methodologies that improve sperm functionality. The first method involved transient exposure to the Ca2+ ionophore A23187 , and the second required sperm incubation in the absence of energy nutrients (starvation). Both methods were associated with an initial loss of motility followed by a rescue step involving ionophore removal or addition of energy metabolites, respectively. In this work, we show that starvation is accompanied by an increase in intracellular Ca2+ ([Ca2+ ]i ). Additionally, the starved cells acquire a significantly enhanced capacity to undergo a progesterone-induced acrosome reaction. Electrophysiological measurements show that CatSper channel remains active in starvation conditions. However, the increase in [Ca2+ ]i was also observed in sperm from CatSper null mice. Upon starvation, addition of energy nutrients reversed the effects on [Ca2+ ]i and decreased the effect of progesterone on the acrosome reaction to control levels. These data indicate that both methods have common molecular features.


Asunto(s)
Calcio/metabolismo , Progesterona/farmacología , Capacitación Espermática/efectos de los fármacos , Inanición/metabolismo , Reacción Acrosómica/efectos de los fármacos , Animales , Canales de Calcio/metabolismo , Membrana Celular/metabolismo , Femenino , Masculino , Ratones , Progesterona/metabolismo , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo
7.
FASEB J ; 35(8): e21723, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34224609

RESUMEN

Sperm acquire the ability to fertilize in a process called capacitation and undergo hyperactivation, a change in the motility pattern, which depends on Ca2+ transport by CatSper channels. CatSper is essential for fertilization and it is subjected to a complex regulation that is not fully understood. Here, we report that similar to CatSper, Cdc42 distribution in the principal piece is confined to four linear domains and this localization is disrupted in CatSper1-null sperm. Cdc42 inhibition impaired CatSper activity and other Ca2+ -dependent downstream events resulting in a severe compromise of the sperm fertilizing potential. We also demonstrate that Cdc42 is essential for CatSper function by modulating cAMP production by soluble adenylate cyclase (sAC), providing a new regulatory mechanism for the stimulation of CatSper by the cAMP-dependent pathway. These results reveal a broad mechanistic insight into the regulation of Ca2+ in mammalian sperm, a matter of critical importance in male infertility as well as in contraception.


Asunto(s)
Canales de Calcio/metabolismo , Espermatozoides/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio/deficiencia , Canales de Calcio/genética , Señalización del Calcio , AMP Cíclico/metabolismo , Femenino , Fertilización In Vitro , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Transducción de Señal , Capacitación Espermática/fisiología , Motilidad Espermática/fisiología , Cola del Espermatozoide/metabolismo , Espermatozoides/efectos de los fármacos , Espermatozoides/ultraestructura , Proteína de Unión al GTP cdc42/antagonistas & inhibidores
8.
Int J Mol Sci ; 24(1)2022 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-36614081

RESUMEN

The sperm energy restriction and recovery (SER) treatment developed in our laboratory was shown to improve fertilization and blastocyst development following in vitro fertilization (IVF) in mice. Here, we investigated the effects of SER on early embryogenesis. Developmental events observed during the first cell cycle indicated that progression through the pronuclear stages of SER-generated embryos is advanced in comparison with control-generated embryos. These findings prompted further analysis of potential effects of SER on pronuclear chromatin dynamics, focusing on the key H3K4me3 and H3K27ac histone modifications. Nearly all the SER-generated embryos displayed H3K4me3 in the male pronuclei at 12 h post-insemination (HPI), while a subset of the control-generated embryos did not. Additionally, SER-generated embryos displayed a more homogenous intensity of H3K27ac at 8 and 12 HPI compared to control embryos. These changes in histone modifications during the first cell cycle were accompanied by differences in gene expression at the two-cell stage; both of these changes in early embryos could potentially play a role in the improved developmental outcomes of these embryos later in development. Our results indicate that sperm incubation conditions have an impact on early embryo development and can be useful for the improvement of assisted reproductive technology outcomes.


Asunto(s)
Fertilización In Vitro , Semen , Masculino , Animales , Ratones , Espermatozoides , Desarrollo Embrionario , Ciclo Celular , Epigénesis Genética , Blastocisto/metabolismo
9.
Physiology (Bethesda) ; 35(3): 196-208, 2020 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-32293232

RESUMEN

Species preservation depends on the success of fertilization. Sperm are uniquely equipped to fulfill this task, and, although several mechanisms are conserved among species, striking functional differences have evolved to contend with particular sperm-egg environmental characteristics. This review highlights similarities and differences in sperm strategies, with examples within internal and external fertilizers, pointing out unresolved issues.


Asunto(s)
Acrosoma , Capacitación Espermática , Humanos , Masculino , Acrosoma/fisiología , Semen , Capacitación Espermática/fisiología , Espermatozoides/fisiología , Animales
10.
Mol Reprod Dev ; 88(8): 544-557, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34318548

RESUMEN

Mammalian sperm have to undergo capacitation to fertilize the egg. At the molecular level, capacitation involves cAMP synthesis, protein kinase A activation, and downstream increase in tyrosine phosphorylation. In addition, during capacitation, mammalian sperm actively generate reactive oxygen species (ROS). It has been proposed that ROS modulate phosphorylation pathways; however, the crosstalk between these signaling processes is not well-understood. In the present study, we used loss- and gain-of-function approaches to evaluate the interconnection between ROS and phosphorylation. We showed that BSA and HCO3- , but not Ca2+ , in the capacitation media are required for ROS production. The synergic effect of these compounds was neither mediated by HCO3- stimulation of cAMP synthesis nor by BSA-induced cholesterol efflux. The capacitation-induced ROS generation was blocked in the presence of superoxide dismutase (SOD), catalase, and apocynin. However, none of these compounds affected cAMP-dependent or tyrosine phosphorylation. On the other hand, the addition of NADPH to the media induced ROS generation in sperm incubated in the absence of BSA and HCO3- without upregulating cAMP-dependent or tyrosine phosphorylation signaling. Most interestingly, catalase, but not SOD, blocked in vitro fertilization suggesting a role for H2 O2 in this process.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Capacitación Espermática/fisiología , Espermatozoides/metabolismo , Animales , Calcio/metabolismo , Catalasa/metabolismo , Masculino , Ratones , Fosforilación , Superóxido Dismutasa/metabolismo
11.
Mol Reprod Dev ; 88(11): 718-730, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34623009

RESUMEN

We have previously shown that members of the family of testis-specific serine/threonine kinases (TSSKs) are post-meiotically expressed in testicular germ cells and in mature sperm in mammals. The restricted post-meiotic expression of TSSKs as well as the importance of phosphorylation in signaling processes strongly suggest that TSSKs have an important role in germ cell differentiation and/or sperm function. This prediction has been supported by the reported sterile phenotype of the TSSK6 knock-out (KO) mice and of the double TSSK1/TSSK2 KO. The aim of this study was to develop KO mouse models of TSSK3 and to validate this kinase as a target for the development of a male contraceptive. We used CRISPR/Cas9 technology to generate the TSSK3 KO allele on B6D2F1 background mice. Male heterozygous pups were used to establish three independent TSSK3 KO lines. After natural mating of TSSK3 KO males, females that presented a plug (indicative of mating) were monitored for the following 24 days and no pregnancies or pups were found. Sperm numbers were drastically reduced in all three KO lines and, remarkably, round spermatids were detected in the cauda epididymis of KO mice. From the small population of sperm recovered, severe morphology defects were detected. Our results indicate an essential role of TSSK3 in spermiogenesis and support this kinase as a suitable candidate for the development of novel nonhormonal male contraceptives.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Espermatogénesis , Testículo , Animales , Anticoncepción , Femenino , Masculino , Mamíferos , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Espermátides , Espermatogénesis/genética , Espermatozoides/metabolismo , Testículo/metabolismo
12.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34638585

RESUMEN

Mammalian sperm must undergo two post-testicular processes to become fertilization-competent: maturation in the male epididymis and capacitation in the female reproductive tract. While caput epididymal sperm are unable to move and have not yet acquired fertilization potential, sperm in the cauda epididymis have completed their maturation, can move actively, and have gained the ability to undergo capacitation in the female tract or in vitro. Due to the impossibility of mimicking sperm maturation in vitro, the molecular pathways underlying this process remain largely unknown. We aimed to investigate the use of caput epididymal ligation as a tool for the study of sperm maturation in mice. Our results indicate that after seven days of ligation, caput sperm gained motility and underwent molecular changes comparable with those observed for cauda mature sperm. Moreover, ligated caput sperm were able to activate pathways related to sperm capacitation. Despite these changes, ligated caput sperm were unable to fertilize in vitro. Our results suggest that transit through the epididymis is not required for the acquisition of motility and some capacitation-associated signaling but is essential for full epididymal maturation. Caput epididymal ligation is a useful tool for the study of the molecular pathways involved in the acquisition of sperm motility during maturation.


Asunto(s)
AMP Cíclico/metabolismo , Fosforilación/fisiología , Maduración del Esperma/fisiología , Motilidad Espermática/fisiología , Espermatozoides/fisiología , Animales , Epidídimo/metabolismo , Epidídimo/fisiología , Femenino , Fertilización/fisiología , Ligadura/métodos , Masculino , Ratones , Transducción de Señal/fisiología , Capacitación Espermática/fisiología , Espermatozoides/metabolismo
13.
J Cell Sci ; 131(11)2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29739876

RESUMEN

Conception in mammals is determined by the fusion of a sperm cell with an oocyte during fertilization. Motility is one of the features of sperm that allows them to succeed in fertilization, and their flagellum is essential for this function. Longitudinally, the flagellum can be divided into the midpiece, the principal piece and the end piece. A precise cytoskeletal architecture of the sperm tail is key for the acquisition of fertilization competence. It has been proposed that the actin cytoskeleton plays essential roles in the regulation of sperm motility; however, the actin organization in sperm remains elusive. In the present work, we show that there are different types of actin structures in the sperm tail by using three-dimensional stochastic optical reconstruction microscopy (STORM). In the principal piece, actin is radially distributed between the axoneme and the plasma membrane. The actin-associated proteins spectrin and adducin are also found in these structures. Strikingly, polymerized actin in the midpiece forms a double-helix that accompanies mitochondria. Our findings illustrate a novel specialized structure of actin filaments in a mammalian cell.


Asunto(s)
Citoesqueleto de Actina/química , Cola del Espermatozoide/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Actinas/química , Actinas/genética , Actinas/metabolismo , Animales , Masculino , Ratones , Conformación Proteica en Hélice alfa , Cola del Espermatozoide/química
14.
J Cell Sci ; 131(21)2018 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-30301778

RESUMEN

Filamentous actin (F-actin) is a key factor in exocytosis in many cell types. In mammalian sperm, acrosomal exocytosis (denoted the acrosome reaction or AR), a special type of controlled secretion, is regulated by multiple signaling pathways and the actin cytoskeleton. However, the dynamic changes of the actin cytoskeleton in live sperm are largely not understood. Here, we used the powerful properties of SiR-actin to examine actin dynamics in live mouse sperm at the onset of the AR. By using a combination of super-resolution microscopy techniques to image sperm loaded with SiR-actin or sperm from transgenic mice containing Lifeact-EGFP, six regions containing F-actin within the sperm head were revealed. The proportion of sperm possessing these structures changed upon capacitation. By performing live-cell imaging experiments, we report that dynamic changes of F-actin during the AR occur in specific regions of the sperm head. While certain F-actin regions undergo depolymerization prior to the initiation of the AR, others remain unaltered or are lost after exocytosis occurs. Our work emphasizes the utility of live-cell nanoscopy, which will undoubtedly impact the search for mechanisms that underlie basic sperm functions.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Acrosoma/metabolismo , Citoesqueleto de Actina/metabolismo , Espermatozoides/metabolismo , Animales , Exocitosis , Masculino , Ratones , Imagen Molecular
15.
Biol Reprod ; 103(4): 791-801, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32614044

RESUMEN

Mammalian sperm are stored in the epididymis in a dormant state. Upon ejaculation, they must immediately start producing sufficient energy to maintain motility and support capacitation. While this increased energy demand during capacitation is well established, it remains unclear how mouse sperm modify their metabolism to meet this need. We now show that capacitating mouse sperm enhance glucose uptake, identifying glucose uptake as a functional marker of capacitation. Using an extracellular flux analyzer, we show that glycolysis and oxidative phosphorylation increase during capacitation. Furthermore, this increase in oxidative phosphorylation is dependent on glycolysis, providing experimental evidence for a link between glycolysis and oxidative phosphorylation in mouse sperm.


Asunto(s)
Metabolismo Energético/fisiología , Capacitación Espermática/fisiología , Espermatozoides/metabolismo , Animales , Supervivencia Celular , Glucosa/metabolismo , Glucólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación Oxidativa , Zona Pelúcida/fisiología
16.
Biol Reprod ; 103(2): 264-274, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32337545

RESUMEN

Male contraception is a very active area of research. Several hormonal agents have entered clinical trials, while potential non-hormonal targets have been brought to light more recently and are at earlier stages of development. The general strategy is to target genes along the molecular pathways of sperm production, maturation, or function, and it is predicted that these novel approaches will hopefully lead to more selective male contraceptive compounds with a decreased side effect burden. Protein kinases are known to play a major role in signaling events associated with sperm differentiation and function. In this review, we focus our analysis on the testis-specific serine kinase (TSSK) protein family. We have previously shown that members of the family of TSSKs are postmeiotically expressed in male germ cells and in mature mammalian sperm. The restricted postmeiotic expression of TSSKs as well as the importance of phosphorylation in signaling processes strongly suggests that TSSKs have an important role in germ cell differentiation and/or sperm function. This prediction has been supported by the reported sterile phenotype of the Tssk6 knockout (KO) mice and of the double Tssk1 and Tssk2 KO mice and by the male subfertile phenotype observed in a Tssk4 KO mouse model.


Asunto(s)
Anticoncepción/métodos , Fertilidad/fisiología , Infertilidad Masculina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Testículo/metabolismo , Animales , Humanos , Infertilidad Masculina/genética , Masculino , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Espermatogénesis/fisiología , Espermatozoides/metabolismo
17.
Mol Reprod Dev ; 87(10): 1037-1047, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32914502

RESUMEN

Mammalian sperm acquire fertilization capacity in the female reproductive tract in a process known as capacitation. During capacitation, sperm change their motility pattern (i.e., hyperactivation) and become competent to undergo the acrosome reaction. We have recently shown that, in the mouse, sperm capacitation is associated with increased uptake of fluorescently labeled deoxyglucose and with extracellular acidification suggesting enhanced glycolysis. Consistently, in the present work we showed that glucose consumption is enhanced in media that support mouse sperm capacitation suggesting upregulation of glucose metabolic pathways. The increase in glucose consumption was modulated by bicarbonate and blocked by protein kinase A and soluble adenylyl cyclase inhibitors. Moreover, permeable cyclic adenosine monophosphate (cAMP) agonists increase glucose consumption in sperm incubated in conditions that do not support capacitation. Also, the increase in glucose consumption was reduced when sperm were incubated in low calcium conditions. Interestingly, this reduction was not overcome with cAMP agonists. Despite these findings, glucose consumption of sperm from Catsper1 knockout mice was similar to the one from wild type suggesting that other sources of calcium are also relevant. Altogether, these results suggest that cAMP and calcium pathways are involved in the regulation of glycolytic energy pathways during murine sperm capacitation.


Asunto(s)
Glucosa/metabolismo , Capacitación Espermática/fisiología , Espermatozoides/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Metabolismo Energético/genética , Glucólisis/fisiología , Masculino , Ratones , Ratones Noqueados , Motilidad Espermática/genética
18.
J Biol Chem ; 293(43): 16830-16841, 2018 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-30213858

RESUMEN

Mammalian sperm must undergo capacitation as a preparation for entering into hyperactivated motility, undergoing the acrosome reaction, and acquiring fertilizing ability. One of the initial capacitation events occurs when sperm encounter an elevated HCO3- concentration. This anion activates the atypical adenylyl cyclase Adcy10, increases intracellular cAMP, and stimulates protein kinase A (PKA). Moreover, an increase in intracellular Ca2+ concentration ([Ca2+] i ) is essential for sperm capacitation. Although a cross-talk between cAMP-dependent pathways and Ca2+ clearly plays an essential role in sperm capacitation, the connection between these signaling events is incompletely understood. Here, using three different approaches, we found that CatSper, the main sperm Ca2+ channel characterized to date, is up-regulated by a cAMP-dependent activation of PKA in mouse sperm. First, HCO3- and the PKA-activating permeable compound 8-Br-cAMP induced an increase in [Ca2+] i , which was blocked by the PKA peptide inhibitor PKI, and H89, another PKA inhibitor, also abrogated the 8-Br-cAMP response. Second, HCO3- increased the membrane depolarization induced upon divalent cation removal by promoting influx of monovalent cations through CatSper channels, which was inhibited by PKI, H89, and the CatSper blocker HC-056456. Third, electrophysiological patch clamp, whole-cell recordings revealed that CatSper activity is up-regulated by HCO3- and by direct cAMP injection through the patch-clamp pipette. The activation by HCO3- and cAMP was also blocked by PKI, H89, Rp-cAMPS, and HC-056456, and electrophysiological recordings in sperm from CatSper-KO mice confirmed CatSper's role in these activation modes. Our results strongly suggest that PKA-dependent phosphorylation regulates [Ca2+] i homeostasis by activating CatSper channel complexes.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Membrana Celular/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Motilidad Espermática/fisiología , Espermatozoides/fisiología , Animales , Canales de Calcio/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Masculino , Ratones , Fosforilación , Capacitación Espermática
19.
J Biol Chem ; 293(24): 9435-9447, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29700114

RESUMEN

Protein kinase A (PKA) is a broad-spectrum Ser/Thr kinase involved in the regulation of several cellular activities. Thus, its precise activation relies on being localized at specific subcellular places known as discrete PKA signalosomes. A-Kinase anchoring proteins (AKAPs) form scaffolding assemblies that play a pivotal role in PKA regulation by restricting its activity to specific microdomains. Because one of the first signaling events observed during mammalian sperm capacitation is PKA activation, understanding how PKA activity is restricted in space and time is crucial to decipher the critical steps of sperm capacitation. Here, we demonstrate that the anchoring of PKA to AKAP is not only necessary but also actively regulated during sperm capacitation. However, we find that once capacitated, the release of PKA from AKAP promotes a sudden Ca2+ influx through the sperm-specific Ca2+ channel CatSper, starting a tail-to-head Ca2+ propagation that triggers the acrosome reaction. Three-dimensional super-resolution imaging confirmed a redistribution of PKA within the flagellar structure throughout the capacitation process, which depends on anchoring to AKAP. These results represent a new signaling event that involves CatSper Ca2+ channels in the acrosome reaction, sensitive to PKA stimulation upon release from AKAP.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Reacción Acrosómica , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Mapas de Interacción de Proteínas , Capacitación Espermática , Espermatozoides/citología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/análisis , Exocitosis , Fertilización , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Espermatozoides/metabolismo
20.
Development ; 143(13): 2325-33, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27226326

RESUMEN

Sperm capacitation is required for fertilization. At the molecular level, this process is associated with fast activation of protein kinase A. Downstream of this event, capacitating conditions lead to an increase in tyrosine phosphorylation. The identity of the tyrosine kinase(s) mediating this process has not been conclusively demonstrated. Recent experiments using stallion and human sperm have suggested a role for PYK2 based on the use of small molecule inhibitors directed against this kinase. However, crucially, loss-of-function experiments have not been reported. Here, we used both pharmacological inhibitors and genetically modified mice models to investigate the identity of the tyrosine kinase(s) mediating the increase in tyrosine phosphorylation in mouse sperm. Similar to stallion and human, PF431396 blocks the capacitation-associated increase in tyrosine phosphorylation. Yet, sperm from Pyk2(-/-) mice displayed a normal increase in tyrosine phosphorylation, implying that PYK2 is not responsible for this phosphorylation process. Here, we show that PF431396 can also inhibit FER, a tyrosine kinase known to be present in sperm. Sperm from mice targeted with a kinase-inactivating mutation in Fer failed to undergo capacitation-associated increases in tyrosine phosphorylation. Although these mice are fertile, their sperm displayed a reduced ability to fertilize metaphase II-arrested eggs in vitro.


Asunto(s)
Fosfotirosina/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Capacitación Espermática/fisiología , Espermatozoides/enzimología , Animales , Quinasa 2 de Adhesión Focal/metabolismo , Masculino , Ratones Endogámicos C57BL , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA