Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Dev Biol ; 487: 24-33, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35439527

RESUMO

The physiological acrosome reaction occurs after mammalian spermatozoa undergo a process called capacitation in the female reproductive tract. Only acrosome reacted spermatozoon can penetrate the egg zona-pellucida and fertilize the egg. Sperm also contain several mechanisms that protect it from undergoing spontaneous acrosome reaction (sAR), a process that can occur in sperm before reaching proximity to the egg and that abrogates fertilization. We previously showed that calmodulin-kinase II (CaMKII) and phospholipase D (PLD) are involved in preventing sAR through two distinct pathways that enhance F-actin formation during capacitation. Here, we describe a novel additional pathway involving the tyrosine kinase Fer in a mechanism that also prevents sAR by enhancing actin polymerization during sperm capacitation. We further show that protein-kinase A (PKA) and the tyrosine-kinase Src, as well as PLD, direct Fer phosphorylation/activation. Activated Fer inhibits the Ser/Thr phosphatase PP1, thereby leading to CaMKII activation, actin polymerization, and sAR inhibition.


Assuntos
Reação Acrossômica , Fosfolipase D , Acrossomo , Reação Acrossômica/fisiologia , Actinas/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Feminino , Masculino , Mamíferos/metabolismo , Proteínas Tirosina Quinases/metabolismo , Capacitação Espermática/fisiologia , Espermatozoides/metabolismo
2.
Int J Mol Sci ; 24(23)2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-38069328

RESUMO

To acquire the capacity to fertilize the oocyte, mammalian spermatozoa must undergo a series of biochemical reactions in the female reproductive tract, which are collectively called capacitation. The capacitated spermatozoa subsequently interact with the oocyte zona-pellucida and undergo the acrosome reaction, which enables the penetration of the oocyte and subsequent fertilization. However, the spontaneous acrosome reaction (sAR) can occur prematurely in the sperm before reaching the oocyte cumulus oophorus, thereby jeopardizing fertilization. One of the main processes in capacitation involves actin polymerization, and the resulting F-actin is subsequently dispersed prior to the acrosome reaction. Several biochemical reactions that occur during sperm capacitation, including actin polymerization, protect sperm from sAR. In the present review, we describe the protective mechanisms that regulate sperm capacitation and prevent sAR.


Assuntos
Reação Acrossômica , Actinas , Animais , Masculino , Feminino , Reação Acrossômica/fisiologia , Sêmen , Espermatozoides/fisiologia , Citoesqueleto de Actina , Mamíferos , Acrossomo
3.
Int J Mol Sci ; 24(6)2023 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-36982326

RESUMO

Fer and its sperm and cancer specific variant, FerT, are non-receptor tyrosine kinases which play roles in cancer progression and metastasis. Recent studies have shed light on the regulatory role of these kinases in ensuring proper sperm function. Comparison of the regulatory cascades in which Fer and FerT are engaged in sperm and cancer cells presents an interesting picture, in which similar regulatory interactions of these enzymes are integrated in a similar or different regulatory context in the two cell types. These diverse compositions extend from the involvement of Fer in modulation of actin cytoskeleton integrity and function, to the unique regulatory interactions of Fer with PARP-1 and the PP1 phosphatase. Furthermore, recent findings link the metabolic regulatory roles of Fer and FerT in sperm and cancer cells. In the current review, we discuss the above detailed aspects, which portray Fer and FerT as new regulatory links between sperm and malignant cells. This perspective view can endow us with new analytical and research tools that will deepen our understanding of the regulatory trajectories and networks that govern these two multi-layered systems.


Assuntos
Neoplasias , Proteínas Tirosina Quinases , Masculino , Humanos , Proteínas Tirosina Quinases/metabolismo , Sêmen/metabolismo , Espermatozoides/metabolismo , Fosforilação , Neoplasias/metabolismo
4.
Int J Mol Sci ; 21(20)2020 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-33096823

RESUMO

Several studies proposed the importance of zinc ion in male fertility. Here, we describe the properties, roles and cellular mechanisms of action of Zn2+ in spermatozoa, focusing on its involvement in sperm motility, capacitation and acrosomal exocytosis, three functions that are crucial for successful fertilization. The impact of zinc supplementation on assisted fertilization techniques is also described. The impact of zinc on sperm motility has been investigated in many vertebrate and invertebrate species. It has been reported that Zn2+ in human seminal plasma decreases sperm motility and that Zn2+ removal enhances motility. Reduction in the intracellular concentration of Zn2+ during epididymal transit allows the development of progressive motility and the subsequent hyper activated motility during sperm capacitation. Extracellular Zn2+ affects intracellular signaling pathways through its interaction with the Zn2+ sensing receptor (ZnR), also named GPR39. This receptor was found in the sperm tail and the acrosome, suggesting the possible involvement of Zn2+ in sperm motility and acrosomal exocytosis. Our studies showed that Zn2+ stimulates bovine sperm acrosomal exocytosis, as well as human sperm hyper-activated motility, were both mediated by GPR39. Zn2+ binds and activates GPR39, which activates the trans-membrane-adenylyl-cyclase (tmAC) to catalyze cAMP production. The NHE (Na+/H+-exchanger) is activated by cAMP, leading in increased pHi and activation of the sperm-specific Ca2+ channel CatSper, resulting in an increase in [Ca2+]i, which, together with HCO3-, activates the soluble adenylyl-cyclase (sAC). The increase in [cAMP]i activates protein kinase A (PKA), followed by activation of the Src-epidermal growth factor receptor-Pphospholipase C (Src-EGFR-PLC) cascade, resulting in inositol-triphosphate (IP3) production, which mobilizes Ca2+ from the acrosome, causing a further increase in [Ca2+]i and the development of hyper-activated motility. PKA also activates phospholipase D1 (PLD1), leading to F-actin formation during capacitation. Prior to the acrosomal exocytosis, PLC induces phosphadidylinositol-4,5-bisphosphate (PIP2) hydrolysis, leading to the release of the actin-severing protein gelsolin to the cytosol, which is activated by Ca2+, resulting in F-actin breakdown and the occurrence of acrosomal exocytosis.


Assuntos
Técnicas de Reprodução Assistida , Espermatozoides/fisiologia , Zinco/metabolismo , Acrossomo/metabolismo , Animais , Fertilidade/fisiologia , Humanos , Masculino , Capacitação Espermática/fisiologia , Motilidade dos Espermatozoides/fisiologia , Zinco/farmacologia
5.
Mol Reprod Dev ; 86(5): 502-515, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30746812

RESUMO

To fertilize the egg, sperm cells must reside in the female reproductive tract for several hours during which they undergo chemical and motility changes collectively called capacitation. During capacitation, the sperm develop a unique type of motility known as hyperactivated motility (HAM). The semen contains Zn2+ in millimolar concentrations, whereas in the female reproductive tract the concentration is around 1 µM. In this study, we characterize the role of Zn 2+ in human sperm capacitation focusing on its effect on HAM. Western blot analysis revealed the presence of G protein-coupled receptor 39 (GPR39) type Zn-receptor localized mainly in the sperm tail. Zn 2+ at micromolar concentration stimulates HAM, which is mediated by a cascade involving GPR39-AC-cAMP-PKA-Src-EGFR and phospholipase C. Both the transmembrane adenylyl cyclase (AC) and the soluble-AC are involved in the stimulation of HAM by Zn 2+ . The development of HAM is precisely regulated by cyclic adenosine monophosphate, in which relatively low concentration (5-10 µM) stimulated HAM, whereas at 30 µM no stimulation occurred. A similar response was seen when different concentrations of Zn 2+ were added to the cells; low Zn 2+ stimulated HAM, whereas at relatively high Zn 2+ , no effect was seen. We further demonstrate that the Ca 2+ -channel CatSper involved in Zn 2+ -stimulated HAM. These data support a role for extracellular Zn 2+ acting via GPR39 to regulate signaling pathways in sperm capacitation, leading to HAM induction.


Assuntos
Transdução de Sinais , Capacitação Espermática , Motilidade dos Espermatozoides , Espermatozoides , Zinco , Adenilil Ciclases/metabolismo , Humanos , Masculino , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Capacitação Espermática/efeitos dos fármacos , Capacitação Espermática/fisiologia , Motilidade dos Espermatozoides/efeitos dos fármacos , Motilidade dos Espermatozoides/fisiologia , Espermatozoides/química , Espermatozoides/efeitos dos fármacos , Espermatozoides/fisiologia , Zinco/metabolismo , Zinco/farmacologia
6.
Biochem Biophys Res Commun ; 506(2): 372-377, 2018 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-29102633

RESUMO

For the acquisition of the ability to fertilize the egg, mammalian spermatozoa should undergo a series of biochemical transformations in the female reproductive tract, collectively called capacitation. The capacitated sperm can undergo the acrosomal exocytosis process near or on the oocyte, which allows the spermatozoon to penetrate and fertilize it. One of the main processes in capacitation involves dynamic cytoskeletal remodeling particularly of actin. Actin polymerization occurs during sperm capacitation and the produced F-actin should be depolymerized prior to the acrosomal exocytosis. In the present review, we describe the mechanisms that regulate F-actin formation during sperm capacitation and the F-actin dispersion prior to the acrosomal exocytosis. During sperm capacitation, the actin severing proteins gelsolin and cofilin are inactive and they undergo activation prior to the acrosomal exocytosis.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Cálcio/metabolismo , Cofilina 1/metabolismo , Gelsolina/metabolismo , Espermatozoides/metabolismo , Reação Acrossômica , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/ultraestrutura , Actinas/química , Actinas/genética , Animais , Cofilina 1/genética , Exocitose , Feminino , Gelsolina/genética , Regulação da Expressão Gênica , Humanos , Masculino , Oócitos/citologia , Oócitos/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Transdução de Sinais , Capacitação Espermática , Espermatozoides/citologia
7.
Mol Reprod Dev ; 85(6): 543-556, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29750435

RESUMO

To fertilize the egg, sperm cells must reside in the female reproductive tract for several hours during which they undergo chemical and motility changes collectively called capacitation. During capacitation, the sperm develop a unique type of motility known as hyperactivated motility (HAM). The semen contains Zn2+ in millimolar concentrations, whereas in the female reproductive tract, the concentration is around 1 µM. In this study, we characterize the role of Zn2+ in human sperm capacitation focusing on its effect on HAM. Western blot analysis revealed the presence of GPR39-type Zn-receptor localized mainly in the sperm tail. Zn2+ at micromolar concentration stimulates HAM, which is mediated by a cascade involving GPR39-adenylyl cyclase (AC)-cyclic AMP (cAMP)-protein kinase A-tyrosine kinase Src (Src)-epidermal growth factor receptor and phospholipase C. Both the transmembrane AC and the soluble-AC are involved in the stimulation of HAM by Zn2+ . The development of HAM is precisely regulated by cAMP, in which relatively low concentration (5-10 µM) stimulated HAM, whereas at 30 µM no stimulation occurred. A similar response was seen when different concentrations of Zn2+ were added to the cells; low Zn2+ stimulated HAM, whereas at relatively high Zn2+ , no effect was seen. We further demonstrate that the Ca2+ -channel CatSper involved in Zn2+ -stimulated HAM. These data support a role for extracellular Zn2+ acting via GPR39 to regulate signaling pathways in sperm capacitation, leading to HAM induction.


Assuntos
Transdução de Sinais/fisiologia , Capacitação Espermática/fisiologia , Motilidade dos Espermatozoides/fisiologia , Espermatozoides/metabolismo , Zinco/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Receptores ErbB/metabolismo , Humanos , Masculino , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Capacitação Espermática/efeitos dos fármacos , Motilidade dos Espermatozoides/efeitos dos fármacos , Espermatozoides/citologia , Zinco/farmacologia
8.
Dev Biol ; 415(1): 64-74, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27178669

RESUMO

In order to interact with the egg and undergo acrosomal exocytosis or the acrosome reaction (AR), mammalian spermatozoa must undergo a series of biochemical changes in the female reproductive tract, collectively called capacitation. We showed that F-actin is formed during sperm capacitation and fast depolymerization occurs prior to the AR. We hypothesized that F-actin protects the sperm from undergoing spontaneous-AR (sAR) which decreases fertilization rate. We show that activation of the actin-severing protein gelsolin induces a significant increase in sAR. Moreover, inhibition of CaMKII or PLD during sperm capacitation, caused an increase in sAR and inhibition of F-actin formation. Spermine, which leads to PLD activation, was able to reverse the effects of CaMKII inhibition on sAR-increase and F-actin-decrease. Furthermore, the increase in sAR and the decrease in F-actin caused by the inactivation of the PLD-pathway, were reversed by activation of CaMKII using H2O2 or by inhibiting protein phosphatase 1 which enhance the phosphorylation and oxidation states of CaMKII. These results indicate that two distinct pathways lead to F-actin formation in the sperm capacitation process which prevents the occurrence of sAR.


Assuntos
Reação Acrossômica/fisiologia , Acrossomo/enzimologia , Actinas/metabolismo , Citoesqueleto de Actina/ultraestrutura , Animais , Calcimicina/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Bovinos , Ativação Enzimática/efeitos dos fármacos , Exocitose/fisiologia , Gelsolina/metabolismo , Gelsolina/farmacologia , Peróxido de Hidrogênio/farmacologia , Masculino , Toxinas Marinhas , Oxazóis/farmacologia , Fragmentos de Peptídeos/farmacologia , Fosfolipase D/antagonistas & inibidores , Fosfolipase D/fisiologia , Polimerização , Capacitação Espermática/fisiologia , Espermatozoides/efeitos dos fármacos , Espermatozoides/ultraestrutura
9.
Mol Reprod Dev ; 84(1): 8-18, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27864906

RESUMO

The serine/threonine kinase Glycogen synthase kinase 3 (GSK-3) is a master switch that regulates a multitude of cellular pathways, including the acrosome reaction in sperm. In epididymal sperm cells, for example, GSK-3 activity correlates with inhibition of motility-yet no direct pathways connecting GSK-3 activation with loss of motility have been described. Indeed, the details of how GSK-3 is regulated during sperm capacitation and the acrosome reaction remains obscure. To this end, we addressed the involvement of the GSK-3 beta isoform in several known pathways that contribute to motility and the acrosome reaction. We established that Protein kinase A (PKA) is the main regulator of GSK-3ß in sperm, as pre-treatment of cells with a GSK-3 inhibitor prior to addition of H89, an inhibitor of PKA, attenuated the motility loss induced by blocking PKA activity. Both induced and spontaneous acrosome reactions also occurred less frequently in sperm treated with GSK-3 inhibitors. Finally, we observed a slow decline in phosphorylation of GSK-3ß on Ser 9, which represents an inhibited state, during sperm capacitation; this phenotype is reversed during the induced acrosome reaction, in parallel to activation of Protein phosphatase 1. These results suggest that maintenance of sperm motility and acrosome reaction timing are mediated by PKA through the regulation of GSK-3 beta activity. Mol. Reprod. Dev. 84: 8-18, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Reação Acrossômica/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Motilidade dos Espermatozoides/fisiologia , Espermatozoides/enzimologia , Animais , Bovinos , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Isoquinolinas/farmacologia , Masculino , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Sulfonamidas/farmacologia
10.
Biochim Biophys Acta ; 1850(9): 1912-20, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26093290

RESUMO

BACKGROUND: The A-kinase anchoring protein (AKAP) family is essential for sperm motility, capacitation and the acrosome reaction. PKA-dependent protein tyrosine phosphorylation occurs in mammalian sperm capacitation including AKAP3. In a recent study, we showed that AKAP3 undergoes degradation under capacitation conditions. Thus, we tested here whether AKAP3 degradation might be regulated by its tyrosine phosphorylation. METHODS: The intracellular levels of AKAP3 were determined by western blot (WB) analysis using specific anti-AKAP3 antibodies. Tyrosine phosphorylation of AKAP3 was tested by immunoprecipitation and WB analysis. Acrosome reaction was examined using FITC-pisum sativum agglutinin. RESULTS: AKAP3 is degraded and undergoes tyrosine-dephosphorylation during sperm capacitation and the degradation was reduced by inhibition of tyrosine phosphatase and enhanced by inhibition of tyrosine kinase. Sperm starvation or inhibition of mitochondrial respiration, which reduce cellular ATP levels, significantly accelerated AKAP3 degradation. Treatment with vanadate, or Na(+) or bicarbonate depletion, reduced AKAP3-degradation and the AR rate, while antimycin A or NH4Cl elevated both AKAP3-degradation and the AR degree. Treatment of sperm with NH4Cl enhanced PKA-dependent phosphorylation of four proteins, further supporting the involvement of AKAP3-degradation in capacitation. To demonstrate more specifically that sperm capacitation requires AKAP3-degradation, we inhibited AKAP3-degradation using anti-AKAP3 antibody in permeabilized cells. The anti-AKAP3-antibody induced significant inhibition of AKAP3-degradation and of the AR rate. CONCLUSION: Sperm capacitation process requires AKAP3-degradation, and the degradation degree is regulated by the level of AKAP3 tyrosine phosphorylation. GENERAL SIGNIFICANCE: Better understanding of the molecular mechanisms that mediate sperm capacitation can be used for infertility diagnosis, treatment and the developing of male contraceptives.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Capacitação Espermática , Tirosina/metabolismo , Animais , Bovinos , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Masculino , Fosforilação
11.
Dev Biol ; 396(2): 246-55, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25446533

RESUMO

Extracellular zinc regulates cell proliferation via the MAP1 kinase pathway in several cell types, and has been shown to act as a signaling molecule. The testis contains a relatively high concentration of Zn(2+), required in both the early and late stages of spermatogenesis. Despite the clinical significance of this ion, its role in mature sperm cells is poorly understood. In this study, we characterized the role of Zn(2+) in sperm capacitation and in the acrosome reaction. Western blot analysis revealed the presence of ZnR of the GPR39 type in sperm cells. We previously demonstrated the presence of active epidermal growth factor receptor (EGFR) in sperm, its possible transactivation by direct activation of G-protein coupled receptor (GPCR), and its involvement in sperm capacitation and in the acrosome reaction (AR). We show here that Zn(2+) activates the EGFR during sperm capacitation, which is mediated by activation of trans-membrane adenylyl cyclase (tmAC), protein kinase A (PKA), and the tyrosine kinase, Src. Moreover, the addition of Zn(2+) to capacitated sperm caused further stimulation of EGFR and phosphatydil-inositol-3-kinase (PI3K) phosphorylation, leading to the AR. The stimulation of the AR by Zn(2+) also occurred in the absence of Ca(2+) in the incubation medium, and required the tmAC, indicating that Zn(2+) activates a GPCR. The AR stimulated by Zn(2+) is mediated by GPR39 receptor, PKA, Src and the EGFR, as well as the EGFR down-stream effectors PI3K, phospholipase C (PLC) and protein kinase C (PKC). These data support a role for extracellular zinc, acting through the ZnR, in regulating multiple signaling pathways in sperm capacitation and the acrosome reaction.


Assuntos
Reação Acrossômica/fisiologia , Receptores ErbB/metabolismo , Transdução de Sinais/fisiologia , Capacitação Espermática/fisiologia , Zinco/farmacologia , Reação Acrossômica/efeitos dos fármacos , Adenilil Ciclases/metabolismo , Animais , Western Blotting , Bovinos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Imuno-Histoquímica , Masculino , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Capacitação Espermática/efeitos dos fármacos , Zinco/administração & dosagem , Quinases da Família src/metabolismo
12.
Biochim Biophys Acta ; 1840(12): 3404-12, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25219457

RESUMO

BACKGROUND: Mitochondria are the major source of ATP to power sperm motility. Phosphorylation of mitochondrial proteins has been proposed as a major regulatory mechanism for mitochondrial bioenergetics. METHODS: Sperm motility was measured by a computer-assisted analyzer, protein detection by western blotting, membrane potential by tetramethylrhodamine, cellular ATP by luciferase assay and localization of PKA by immuno-electron microscopy. RESULTS: Bicarbonate is essential for the creation of mitochondrial electro-chemical gradient, ATP synthesis and sperm motility. Bicarbonate stimulates PKA-dependent phosphorylation of two 60kDa proteins identified as Tektin and glucose-6-phosphate isomerase. This phosphorylation was inhibited by respiration inhibition and phosphorylation could be restored by glucose in the presence of bicarbonate. However, this effect of glucose cannot be seen when the mitochondrial ATP/ADP exchanger was inhibited indicating that glycolytic-produced ATP is transported into the mitochondria and allows PKA-dependent protein phosphorylation inside the mitochondria. CONCLUSIONS: Bicarbonate activates mitochondrial soluble adenylyl cyclase (sAC) which catalyzes cAMP production leading to the activation of mitochondrial PKA. Glucose can overcome the lack of ATP in the absence of bicarbonate but it cannot affect the mitochondrial sAC/PKA system, therefore the PKA-dependent phosphorylation of the 60kDa proteins does not occur in the absence of bicarbonate. GENERAL SIGNIFICANCE: Production of CO2 in Krebs cycle, which is converted to bicarbonate is essential for sAC/PKA activation leading to mitochondrial membrane potential creation and ATP synthesis.

13.
Cell Tissue Res ; 362(3): 665-75, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26085344

RESUMO

The spermatozoon is capable of fertilizing an oocyte only after undergoing several biochemical changes in the female reproductive tract, referred to as capacitation. The capacitated spermatozoon interacts with the egg zona pellucida and undergoes the acrosome reaction, which enables its penetration into the egg and fertilization. Actin dynamics play a major role throughout all these processes. Actin polymerization occurs during capacitation, whereas prior to the acrosome reaction, F-actin must undergo depolymerization. In the present study, we describe the presence of the actin-severing protein, cofilin, in human sperm. We examined the function and regulation of cofilin during human sperm capacitation and compared it to gelsolin, an actin-severing protein that was previously investigated by our group. In contrast to gelsolin, we found that cofilin is mainly phosphorylated/inhibited at the beginning of capacitation, and dephosphorylation occurs towards the end of the process. In addition, unlike gelsolin, cofilin phosphorylation is not affected by changing the cellular levels of PIP2. Despite the different regulation of the two proteins, the role of cofilin appears similar to that of gelsolin, and its activation leads to actin depolymerization, inhibition of sperm motility and induction of the acrosome reaction. Moreover, like gelsolin, cofilin translocates from the tail to the head during capacitation. In summary, gelsolin and cofilin play a similar role in F-actin depolymerization prior to the acrosome reaction but their pattern of phosphorylation/inactivation during the capacitation process is different. Thus, for the sperm to achieve high levels of F-actin along the capacitation process, both proteins must be inactivated at different times and, in order to depolymerize F-actin, both must be activated prior to the acrosome reaction.


Assuntos
Reação Acrossômica , Fatores de Despolimerização de Actina/metabolismo , Capacitação Espermática , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Reação Acrossômica/efeitos dos fármacos , Actinas/metabolismo , Benzoquinonas/farmacologia , Humanos , Isoquinolinas/farmacologia , Lactamas Macrocíclicas/farmacologia , Masculino , Fosfatidilinositol 4,5-Difosfato , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Capacitação Espermática/efeitos dos fármacos , Motilidade dos Espermatozoides/efeitos dos fármacos , Sulfonamidas/farmacologia
14.
Zygote ; 23(5): 652-61, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25023718

RESUMO

Mammalian sperm acquire fertilization capacity after residing in the female reproductive tract for a few hours in a process called capacitation. Only capacitated sperm can bind the zona pellucida (ZP) of the egg and undergo the acrosome reaction, a process that allows penetration and fertilization. Extracellular signal regulated kinase (ERK1/2) mediates signalling in many cell types, however its role in sperm function is largely unknown. Here we show that ERK1/2 is highly phosphorylated/activated after a short incubation of mouse sperm under capacitation conditions and that this phosphorylation is reduced after longer incubation. Further phosphorylation was observed upon addition of crude extract of egg ZP or epidermal growth factor (EGF). The mitogen-activated ERK-kinase (MEK) inhibitor U0126 abolished ERK1/2 phosphorylation, in vitro fertilization rate and the acrosome reaction induced by ZP or EGF but not by the Ca2+-ionophore A23187. Moreover, inhibition of ERK1/2 along the capacitation process diminished almost completely the sperm's ability to go through the acrosome reaction, while inhibition at the end of capacitation attenuated the acrosome reaction rate by only 45%. The fact that the acrosome reaction, induced by the Ca2+ -ionophore A23187, was not inhibited by U0126 suggests that ERK1/2 mediates the acrosome reaction by activating Ca2+ transport into the cell. Direct determination of intracellular [Ca2+] revealed that Ca2+ influx induced by EGF or ZP was completely blocked by U0126. Thus, it has been established that the increase in ERK1/2 phosphorylation/activation in response to ZP or by activation of the EGF receptor (EGFR) by EGF, is a key event for intracellular Ca2+ elevation and the subsequent occurrence of the acrosome reaction.


Assuntos
Reação Acrossômica/efeitos dos fármacos , Cálcio/metabolismo , Inibidores Enzimáticos/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Capacitação Espermática/fisiologia , Espermatozoides/metabolismo , Acrossomo , Animais , Citoplasma/metabolismo , Feminino , Fertilização/efeitos dos fármacos , Immunoblotting , Técnicas Imunoenzimáticas , Masculino , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Capacitação Espermática/efeitos dos fármacos , Interações Espermatozoide-Óvulo/efeitos dos fármacos , Espermatozoides/citologia , Espermatozoides/efeitos dos fármacos , Zona Pelúcida/efeitos dos fármacos , Zona Pelúcida/metabolismo
15.
Dev Biol ; 381(1): 62-72, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23791551

RESUMO

Actin polymerization and development of hyperactivated (HA) motility are two processes that take place during sperm capacitation. In previous studies, we demonstrated that the increase in F-actin during capacitation depends upon inactivation of the actin severing protein, gelsolin, by its binding to phosphatydilinositol-4, 5-bisphosphate (PIP2). Here, we showed for the first time the involvement of PIP2/gelsolin in human sperm motility before and during capacitation. Activation of gelsolin by causing its release from PIP2 inhibited sperm motility, which could be restored by adding PIP2 to the cells. Reduction of PIP2 synthesis inhibited actin polymerization and motility, and increasing PIP2 synthesis enhanced these activities. Furthermore, sperm demonstrating low motility contained low levels of PIP2 and F-actin. During capacitation there was an increase in PIP2 and F-actin levels in the sperm head and a decrease in the tail. In sperm with high motility, gelsolin was mainly localized to the sperm head before capacitation, whereas in low motility sperm, most of the gelsolin was localized to the tail before capacitation and translocated to the head during capacitation. We also showed that phosphorylation of gelsolin on tyrosine-438 depends on its binding to PIP2. Activation of phospholipase C by Ca(2+)-ionophore or by activating the epidermal-growth-factor-receptor inhibits tyrosine phosphorylation of gelsolin. In conclusion, the data indicate that the increase of PIP2 and/or F-actin in the head during capacitation enhances gelsolin translocation to the head. As a result the decrease of gelsolin in the tail allows keeping high level of F-actin in the tail, which is essential for the development of HA motility.


Assuntos
Actinas/química , Fosfatidilinositol 4,5-Difosfato/farmacologia , Motilidade dos Espermatozoides/efeitos dos fármacos , Reação Acrossômica , Cálcio/química , Movimento Celular , Inibidores Enzimáticos/farmacologia , Receptores ErbB/metabolismo , Gelsolina/química , Humanos , Ionóforos/química , Masculino , Polimerização , Capacitação Espermática , Espermatozoides/metabolismo , Fosfolipases Tipo C/química , Tirosina/química
16.
Dev Biol ; 362(2): 154-61, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22178154

RESUMO

In order to fertilize the oocyte, sperm must undergo a series of biochemical changes in the female reproductive tract, known as capacitation. Once capacitated, spermatozoon can bind to the zona pellucida of the egg and undergo the acrosome reaction (AR), a process that enables its penetration and fertilization of the oocyte. Important processes that characterize sperm capacitation are actin polymerization and the development of hyper-activated motility (HAM). Previously, we showed that Phospholipase D (PLD)-dependent actin polymerization occurs during sperm capacitation, however the role of this process in sperm capacitation is not yet known. In the present study, we showed for the first time the involvement of PLD-dependent actin polymerization in sperm motility during mouse and human capacitation. Sperm incubated under capacitation conditions revealed a time dependent increase in actin polymerization and HAM. Inhibition of Phosphatidic Acid (PA) formation by PLD using butan-1-ol, inhibited actin polymerization and motility, as well as in vitro fertilization (IVF) and the ability of the sperm to undergo the AR. The inhibition of sperm HAM by low concentration of butan-1-ol is completely restored by adding PA, further indicating the involvement of PLD in these processes. Furthermore, exogenous PA enhanced rapid actin polymerization that was followed by a rise in the HAM, as well as an increased in IVF rate. In conclusion, our results demonstrate that PLD-dependent actin polymerization is a critical step needed for the development of HAM during mouse and human sperm capacitation.


Assuntos
Reação Acrossômica/fisiologia , Actinas/metabolismo , Fosfolipase D/metabolismo , Capacitação Espermática/fisiologia , Motilidade dos Espermatozoides/fisiologia , Zona Pelúcida/metabolismo , 1-Butanol/farmacologia , Animais , Feminino , Fertilização in vitro , Humanos , Immunoblotting , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Ácidos Fosfatídicos/antagonistas & inibidores , Ácidos Fosfatídicos/metabolismo , Polimerização/efeitos dos fármacos , Motilidade dos Espermatozoides/efeitos dos fármacos
17.
J Biol Chem ; 287(26): 22328-40, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22577141

RESUMO

To attain fertilization the spermatozoon binds to the egg zona pellucida (ZP) via sperm receptor(s) and undergoes an acrosome reaction (AR). Several sperm receptors have been described in the literature; however, the identity of this receptor is not yet certain. In this study, we suggest that the α7 nicotinic acetylcholine receptor (α7nAChR) might be a sperm receptor activated by ZP to induce epidermal growth factor receptor (EGFR)-mediated AR. We found that isolated ZP or α7 agonists induced the AR in sperm from WT but not α7-null spermatozoa, and the induced AR was inhibited by α7 or EGFR antagonists. Moreover, α7-null sperm showed very little binding to the egg, and microfluidic affinity in vitro assay clearly showed that α7nAChR, as well as EGFR, interacted with ZP3. Induction of EGFR activation and the AR by an α7 agonist was inhibited by a Src family kinase (SFK) inhibitor. In conclusion we suggest that activation of α7 by ZP leads to SFK-dependent EGFR activation, Ca(2+) influx, and the acrosome reaction.


Assuntos
Reação Acrossômica , Receptores ErbB/metabolismo , Receptores Nicotínicos/química , Espermatozoides/metabolismo , Quinases da Família src/metabolismo , Animais , Feminino , Fertilização , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas Analíticas Microfluídicas , Reação em Cadeia da Polimerase/métodos , Ligação Proteica , Receptores Nicotínicos/metabolismo , Zona Pelúcida , Receptor Nicotínico de Acetilcolina alfa7
18.
Theriogenology ; 203: 82-88, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36989544

RESUMO

Bacterial contamination in the semen deteriorates spermatozoa function. One mechanism through which this may occur is by inducing a premature form of the acrosome reaction (spontaneous acrosome reaction (sAR)) which has been shown to abrogate fertilization. To understand the mechanism by which bacteria affect sperm functions, we determined the effects of bacteria on sperm sAR and on other parameters involved in sperm capacitation. Sperm cells undergo biochemical changes in the female reproductive tract collectively called capacitation. Only capacitated sperm can undergo the physiological acrosomal exocytosis process near or on the oocyte, which allows the spermatozoon to penetrate and fertilize the egg. Bovine sperm incubated with the bacteria Escherichia coli (E. coli), Staphylococcus aureus (S. aureus) or Pseudomonas aeruginosa (P. aeruginosa), revealed a sperm-bacteria interaction, however only E. coli and P. aeruginosa caused an increase in sperm sAR. This effect was seen only when the bacteria were present with the sperm during the full incubation under capacitation conditions but not when the bacteria were added to capacitated sperm. These results indicate that bacteria affect sperm during capacitation and not at the AR step. In addition, Ca2+ influx, protein kinase A, and protein tyrosine phosphorylation activities, three essential processes that promote capacitation, were inhibited by the bacteria. Moreover, increasing intracellular cAMP, which also occur during sperm capacitation, caused significant reverse of sAR induced by the bacteria.


Assuntos
Reação Acrossômica , Sêmen , Masculino , Bovinos , Animais , Feminino , Escherichia coli , Staphylococcus aureus , Espermatozoides , Capacitação Espermática , Acrossomo/fisiologia
19.
Theriogenology ; 192: 38-44, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-36044805

RESUMO

In order to fertilize the egg, spermatozoa must undergo a series of biochemical processes in the female reproductive tract collectively called capacitation. Only capacitated sperm can interact with the egg resulting in the acrosome reaction (AR), allowing egg penetration and fertilization. Sperm can undergo spontaneous AR (sAR) before reaching the egg, preventing successful fertilization. Here we investigated the metabolic pathways involved in sperm capacitation and sAR. Inhibition of glycolysis or oxidative phosphorylation did not affect capacitation or sAR levels; however, when both systems were inhibited, no capacitation occurred, and there was a significant increase in sAR. Under such ATP-starvation, the increase in sAR is triggered by Ca2+ influx into the sperm via the CatSper cation channel. Protein kinase A (PKA) is an essential key enzyme in sperm capacitation; there was no change in its activity when a single metabolic system was inhibited, while complete inhibition of was observed when the two systems were inhibited. Protein tyrosine phosphorylation (PTP), also known to occur in sperm capacitation, was partially reduced by inhibition of one metabolic system, and completely blocked when the two metabolic systems were inhibited. We conclude that ATP, PKA and PTP are involved in the mechanisms protecting sperm from sAR.


Assuntos
Reação Acrossômica , Sêmen , Acrossomo/metabolismo , Reação Acrossômica/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Masculino , Redes e Vias Metabólicas , Sêmen/metabolismo , Capacitação Espermática/fisiologia , Espermatozoides/fisiologia , Tirosina/metabolismo
20.
Dev Biol ; 344(2): 650-7, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20580701

RESUMO

The sperm acrosome reaction occurs after the binding of the capacitated sperm to the egg zona pellucida. This study describes a novel mode of regulation of the sperm epidermal growth factor receptor (EGFR) under physiological conditions and its relevance to the acrosome reaction. Ouabain, a known Na/K ATPase blocker is present in the blood and in the female reproductive tract. We show here that physiological concentrations (nM) of ouabain enhance phosphorylation of EGFR on tyr-845, stimulate Ca(2+) influx and induce the acrosome reaction in sperm. These effects could be seen only in the presence of very low concentrations of EGF (0.1 ng/ml or 0.016 nM) added together with nano-molar ouabain. Phosphorylation, Ca(2+) influx, and the acrosome reaction are inhibited by an EGFR blocker, suggesting that trans-activation of the EGFR is involved. Moreover, our data revealed that protein kinase A and the family of tyrosine kinase, SRC, shown before to be involved in EGFR activation in sperm, mediate the acrosome reaction induced by ouabain. Ouabain alone (without EGF) at relatively high concentration (10microM) could enhance EGFR phosphorylation, Ca(2+) influx and acrosome reaction, and these processes were inhibited by EGFR blockers. Moreover, we show here that PKA and SRC family are involved in the activation of EGFR by 10 microM ouabain, further demonstrating that ouabain induces the acrosome reaction by a mechanism mediated by the trans-activation of EGFR. In conclusion, this study describes an interesting regulatory path of EGFR by physiological concentrations of ouabain and EGF found in the female reproductive tract. Neither of these compounds can activate the EGFR alone at such low physiological levels; however, when both are present, the interaction of ouabain with the Na/K ATPase leads to the priming of the EGFR, which undergoes its full activation by EGF.


Assuntos
Reação Acrossômica/efeitos dos fármacos , Receptores ErbB/metabolismo , Receptores ErbB/fisiologia , Ouabaína/metabolismo , Espermatozoides/fisiologia , Animais , Bovinos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/farmacologia , Feminino , Masculino , Ouabaína/farmacologia , Óvulo/metabolismo , Fosforilação , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas Tirosina Quinases/farmacologia , Receptores de Superfície Celular , Capacitação Espermática/efeitos dos fármacos , Capacitação Espermática/fisiologia , Espermatozoides/metabolismo , Transativadores/metabolismo , Transativadores/farmacologia , Transcrição Gênica/efeitos dos fármacos , Zona Pelúcida/metabolismo , Zona Pelúcida/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA