Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 11: 1148831, 2023.
Article in English | MEDLINE | ID: mdl-37065849

ABSTRACT

Before fertilization, spermatozoa must undergo calcium-regulated acrosome exocytosis in response to physiological stimuli such as progesterone and zona pellucida. Our laboratory has elucidated the signaling cascades accomplished by different sphingolipids during human sperm acrosomal exocytosis. Recently, we established that ceramide increases intracellular calcium by activating various channels and stimulating the acrosome reaction. However, whether ceramide induces exocytosis on its own, activation of the ceramide kinase/ceramide 1-phosphate (CERK/C1P) pathway or both is still an unsolved issue. Here, we demonstrate that C1P addition induces exocytosis in intact, capacitated human sperm. Real-time imaging in single-cell and calcium measurements in sperm population showed that C1P needs extracellular calcium to induce [Ca2+]i increase. The sphingolipid triggered the cation influx through voltage-operated calcium (VOC) and store-operated calcium (SOC) channels. However, it requires calcium efflux from internal stores through inositol 3-phosphate receptors (IP3R) and ryanodine receptors (RyR) to achieve calcium rise and the acrosome reaction. We report the presence of the CERK in human spermatozoa, the enzyme that catalyzes C1P synthesis. Furthermore, CERK exhibited calcium-stimulated enzymatic activity during the acrosome reaction. Exocytosis assays using a CERK inhibitor demonstrated that ceramide induces acrosomal exocytosis, mainly due to C1P synthesis. Strikingly, progesterone required CERK activity to induce intracellular calcium increase and acrosome exocytosis. This is the first report, implicating the bioactive sphingolipid C1P in the physiological progesterone pathway leading to the sperm acrosome reaction.

2.
Nat Commun ; 12(1): 3855, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34158477

ABSTRACT

Human voltage-gated proton channels (hHv1) extrude protons from cells to compensate for charge and osmotic imbalances due metabolism, normalizing intracellular pH and regulating protein function. Human albumin (Alb), present at various levels throughout the body, regulates oncotic pressure and transports ligands. Here, we report Alb is required to activate hHv1 in sperm and neutrophils. Dose-response studies reveal the concentration of Alb in semen is too low to activate hHv1 in sperm whereas the higher level in uterine fluid yields proton efflux, allowing capacitation, the acrosomal reaction, and oocyte fertilization. Likewise, Alb activation of hHv1 in neutrophils is required to sustain production and release of reactive oxygen species during the immune respiratory burst. One Alb binds to both voltage sensor domains (VSDs) in hHv1, enhancing open probability and increasing proton current. A computational model of the Alb-hHv1 complex, validated by experiments, identifies two sites in Alb domain II that interact with the VSDs, suggesting an electrostatic gating modification mechanism favoring the active "up" sensor conformation. This report shows how sperm are triggered to fertilize, resolving how hHv1 opens at negative membrane potentials in sperm, and describes a role for Alb in physiology that will operate in the many tissues expressing hHv1.


Subject(s)
Albumins/metabolism , Inflammation Mediators/metabolism , Ion Channels/metabolism , Neutrophils/metabolism , Sperm Capacitation/physiology , Acrosome Reaction/physiology , Albumins/chemistry , Amino Acid Sequence , Fertilization/physiology , Humans , Ion Channel Gating/physiology , Ion Channels/chemistry , Ion Channels/genetics , Male , Membrane Potentials/physiology , Molecular Dynamics Simulation , Protein Binding , Protein Domains , Protons , Semen/cytology , Semen/metabolism , Sequence Homology, Amino Acid , Spermatozoa/physiology , Static Electricity
3.
Andrology ; 9(4): 1227-1241, 2021 07.
Article in English | MEDLINE | ID: mdl-33609309

ABSTRACT

BACKGROUND: The signaling pathways of the intracellular second messengers cAMP and Ca2+ play a crucial role in numerous physiological processes in human spermatozoa. One such process is the acrosome reaction (AR), which is necessary for spermatozoa to traverse the egg envelope and to expose a fusogenic membrane allowing the egg-sperm fusion. Progesterone and zona pellucida elicit an intracellular Ca2+ increase that is needed for the AR in the mammalian spermatozoa. This increase is mediated by an initial Ca2+ influx but also by a release from intracellular Ca2+ stores. It is known that intracellular Ca2+ stores play a central role in the regulation of [Ca2+ ]i and in the generation of complex Ca2+ signals such as oscillations and waves. In the human spermatozoa, it has been proposed that the cAMP analog and specific agonist of Epac 8-(p-chlorophenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate (2'-O-Me-cAMP) elicits an intracellular Ca2+ release involved in the AR. OBJECTIVE: To identify the molecular entities involved in the Ca2+ mobilization triggered by 2'-O-Me-cAMP in human spermatozoa. MATERIALS AND METHODS: In capacitated human spermatozoa, we monitored Ca2+ dynamics and the occurrence of the AR in real time using Fluo 3-AM and FM4-64 in a Ca2+ -free medium. RESULTS: Epac activation by 2'-O-Me-cAMP induced a Ca2+ wave that started in the midpiece and propagated to the acrosome region. This Ca2+ response was sensitive to rotenone, CGP, xestospongin, NED-19, and thapsigargin, suggesting the participation of different ion transporters (mitochondrial complex I and Na+ /Ca2+ exchanger, inositol 3-phosphate receptors, two-pore channels and internal store Ca2+ -ATPases). DISCUSSION: Our results suggest that Epac activation promotes a dynamic crosstalk between three different intracellular Ca2+ stores: the mitochondria, the redundant nuclear envelope, and the acrosome. CONCLUSION: The Ca2+ wave triggered by Epac activation is necessary to induce the AR and to enhance the flagellar beat.


Subject(s)
Acrosome Reaction/physiology , Calcium Signaling/physiology , Guanine Nucleotide Exchange Factors/metabolism , Spermatozoa/metabolism , Humans , Male
4.
Front Cell Dev Biol ; 8: 575126, 2020.
Article in English | MEDLINE | ID: mdl-33102481

ABSTRACT

Mammalian sperm acquire ability to fertilize through a process called capacitation, occurring after ejaculation and regulated by both female molecules and male decapacitation factors. Bicarbonate and calcium present in the female reproductive tract trigger capacitation in sperm, leading to acrosomal responsiveness and hyperactivated motility. Male decapacitating factors present in the semen avert premature capacitation, until detached from the sperm surface. However, their mechanism of action remains elusive. Here we describe for the first time the molecular basis for the decapacitating action of the seminal protein SPINK3 in mouse sperm. When present in the capacitating medium, SPINK3 inhibited Src kinase, a modulator of the potassium channel responsible for plasma membrane hyperpolarization. Lack of hyperpolarization affected calcium channels activity, impairing the acquisition of acrosomal responsiveness and blocking hyperactivation. Interestingly, SPINK3 acted only on non-capacitated sperm, as it did not bind to capacitated cells. Binding selectivity allows its decapacitating action only in non-capacitated sperm, without affecting capacitated cells.

5.
Proc Natl Acad Sci U S A ; 115(50): E11847-E11856, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30478045

ABSTRACT

Using a de novo peptide inhibitor, Corza6 (C6), we demonstrate that the human voltage-gated proton channel (hHv1) is the main pathway for H+ efflux that allows capacitation in sperm and permits sustained reactive oxygen species (ROS) production in white blood cells (WBCs). C6 was identified by a phage-display strategy whereby ∼1 million novel peptides were fabricated on an inhibitor cysteine knot (ICK) scaffold and sorting on purified hHv1 protein. Two C6 peptides bind to each dimeric channel, one on the S3-S4 loop of each voltage sensor domain (VSD). Binding is cooperative with an equilibrium affinity (Kd) of ∼1 nM at -50 mV. As expected for a VSD-directed toxin, C6 inhibits by shifting hHv1 activation to more positive voltages, slowing opening and speeding closure, effects that diminish with membrane depolarization.


Subject(s)
Ion Channels/physiology , Leukocytes/metabolism , Sperm Capacitation/physiology , Acrosome Reaction/drug effects , Acrosome Reaction/physiology , Amino Acid Sequence , Binding Sites , HEK293 Cells , Humans , Ion Channels/antagonists & inhibitors , Ion Channels/genetics , Male , Membrane Potentials , Peptide Library , Peptides/chemistry , Peptides/pharmacology , Reactive Oxygen Species/metabolism , Respiratory Burst , Sperm Capacitation/drug effects , Toxins, Biological/chemistry , Toxins, Biological/pharmacology
6.
Biol Open ; 7(12)2018 Dec 03.
Article in English | MEDLINE | ID: mdl-30341105

ABSTRACT

Oocyte in vitro maturation does not entirely support all the nuclear and cytoplasmic changes that occur physiologically, and it is poorly understood whether in vitro maturation affects the competence of cortical granules to secrete their content during cortical reaction. Here, we characterize cortical granule exocytosis (CGE) in live mouse oocytes activated by strontium chloride using the fluorescent lectin FITC-LCA. We compared the kinetic of CGE between ovulated (in vivo matured, IVO) and in vitro matured (IVM) mouse oocytes. Results show that: (1) IVM oocytes have a severely reduced response to strontium chloride; (2) the low response was confirmed by quantification of remnant cortical granules in permeabilized cells and by a novel method to quantify the exudate in non-permeabilized cells; (3) the kinetic of CGE in IVO oocytes was rapid and synchronous; (4) the kinetic of CGE in IVM oocytes was delayed and asynchronous; (5) cortical granules in IVM oocytes show an irregular limit in regards to the cortical granule free domain. We propose the analysis of CGE in live oocytes as a biological test to evaluate the competence of IVM mouse oocytes.This article has an associated First Person interview with the first author of the paper.

7.
J Biol Chem ; 290(15): 9823-41, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25713146

ABSTRACT

Regulated secretion is a central issue for the specific function of many cells; for instance, mammalian sperm acrosomal exocytosis is essential for egg fertilization. ARF6 (ADP-ribosylation factor 6) is a small GTPase implicated in exocytosis, but its downstream effectors remain elusive in this process. We combined biochemical, functional, and microscopy-based methods to show that ARF6 is present in human sperm, localizes to the acrosomal region, and is required for calcium and diacylglycerol-induced exocytosis. Results from pulldown assays show that ARF6 exchanges GDP for GTP in sperm challenged with different exocytic stimuli. Myristoylated and guanosine 5'-3-O-(thio)triphosphate (GTPγS)-loaded ARF6 (active form) added to permeabilized sperm induces acrosome exocytosis even in the absence of extracellular calcium. We explore the ARF6 signaling cascade that promotes secretion. We demonstrate that ARF6 stimulates a sperm phospholipase D activity to produce phosphatidic acid and boosts the synthesis of phosphatidylinositol 4,5-bisphosphate. We present direct evidence showing that active ARF6 increases phospholipase C activity, causing phosphatidylinositol 4,5-bisphosphate hydrolysis and inositol 1,4,5-trisphosphate-dependent intra-acrosomal calcium release. We show that active ARF6 increases the exchange of GDP for GTP on Rab3A, a prerequisite for secretion. We propose that exocytic stimuli activate ARF6, which is required for acrosomal calcium efflux and the assembly of the membrane fusion machinery. This report highlights the physiological importance of ARF6 as a key factor for human sperm exocytosis and fertilization.


Subject(s)
ADP-Ribosylation Factors/metabolism , Acrosome/physiology , Exocytosis/physiology , Lipid Metabolism/physiology , rab3A GTP-Binding Protein/metabolism , ADP-Ribosylation Factor 6 , Acrosome/drug effects , Acrosome Reaction/drug effects , Acrosome Reaction/physiology , Calcium/metabolism , Cells, Cultured , Diglycerides/pharmacology , Exocytosis/drug effects , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Immunoblotting , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Male , Microscopy, Confocal , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase D/metabolism , Spermatozoa/drug effects , Spermatozoa/metabolism , Spermatozoa/physiology , Type C Phospholipases/metabolism
8.
Fertil Steril ; 99(1): 99-106.e2, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22999796

ABSTRACT

OBJECTIVE: To evaluate perfringolysin O, a cholesterol-dependent pore-forming cytolysin, as a tool to study several aspects of human sperm physiology. DESIGN: Prospective study. SETTING: Basic research laboratory. PATIENT(S): Human semen samples with normal parameters obtained from healthy donors. INTERVENTION(S): Interaction of recombinant perfringolysin O with human spermatozoa. MAIN OUTCOME MEASURE(S): Assessment of perfringolysin O binding to spermatozoa, tests for acrosome and plasma membrane integrity, and acrosomal exocytosis assays. RESULT(S): Perfringolysin O associated with human spermatozoa at 4°C. The binding was sensitive to changes in cholesterol concentrations and distribution occurring in the plasma membrane of these cells during capacitation. When perfringolysin O-treated sperm were incubated at 37°C, the plasma membrane became permeable, whereas the acrosome membrane remained intact. Permeabilized spermatozoa were able to respond to exocytic stimuli. The process was inhibited by proteins that interfere with membrane fusion, indicating that large molecules, including antibodies, were able to permeate into the spermatozoa. CONCLUSION(S): PFO is a useful probe to assess changes in the amount and distribution of the active sterol fraction present in the sperm plasma membrane. The toxin can be used for the efficient and selective permeabilization of this membrane, rendering a flexible experimental model suitable for studying molecular processes occurring in the sperm cytoplasm.


Subject(s)
Bacterial Toxins/pharmacology , Hemolysin Proteins/pharmacology , Perforin/pharmacology , Spermatozoa/drug effects , Spermatozoa/physiology , Acrosome/drug effects , Acrosome/physiology , Cell Membrane/drug effects , Cell Membrane/physiology , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Exocytosis/drug effects , Humans , In Vitro Techniques , Male , Prospective Studies , Sperm Capacitation/drug effects , Sperm Capacitation/physiology
9.
Biochim Biophys Acta ; 1821(9): 1186-99, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22609963

ABSTRACT

Acrosomal exocytosis involves a massive fusion between the outer acrosomal and the plasma membranes of the spermatozoon triggered by stimuli that open calcium channels at the plasma membrane. Diacylglycerol has been implicated in the activation of these calcium channels. Here we report that this lipid promotes the efflux of intraacrosomal calcium and triggers exocytosis in permeabilized human sperm, implying that diacylglycerol activates events downstream of the opening of plasma membrane channels. Furthermore, we show that calcium and diacylglycerol converge in a signaling pathway leading to the production of phosphatidylinositol 4,5-bisphosphate (PIP(2)). Addition of diacylglycerol promotes the PKC-dependent activation of PLD1. Rescue experiments adding phosphatidic acid or PIP(2) and direct measurement of lipid production suggest that both PKC and PLD1 promote PIP(2) synthesis. Inhibition of different steps of the pathway was reverted by adenophostin, an agonist of IP(3)-sensitive calcium channels, indicating that PIP(2) is necessary to keep these channels opened. However, phosphatidic acid, PIP(2), or adenophostin could not trigger exocytosis by themselves, indicating that diacylglycerol must also activate another factor. We found that diacylglycerol and phorbol ester stimulate the accumulation of the GTP-bound form of Rab3A. Together our results indicate that diacylglycerol promotes acrosomal exocytosis by i) maintaining high levels of IP(3) - an effect that depends on a positive feedback loop leading to the production of PIP(2) - and ii) stimulating the activation of Rab3A, which in turn initiates a cascade of protein interactions leading to the assembly of SNARE complexes and membrane fusion.


Subject(s)
Acrosome/metabolism , Diglycerides/pharmacology , Exocytosis/drug effects , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase D/metabolism , Protein Kinase C/metabolism , Signal Transduction/drug effects , Calcium Channel Agonists/pharmacology , Calcium Channels/metabolism , Exocytosis/physiology , Humans , Male , Membrane Fusion/drug effects , Membrane Fusion/physiology , SNARE Proteins/metabolism , Signal Transduction/physiology , rab3A GTP-Binding Protein/metabolism
10.
Asian J Androl ; 13(1): 159-65, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20835262

ABSTRACT

The acrosome reaction (AR), an absolute requirement for spermatozoa and egg fusion, requires the influx of Ca²(+) into the spermatozoa through voltage-dependent Ca²(+) channels and store-operated channels. Maitotoxin (MTx), a Ca²(+)-mobilizing agent, has been shown to be a potent inducer of the mouse sperm AR, with a pharmacology similar to that of the zona pellucida (ZP), possibly suggesting a common pathway for both inducers. Using recombinant human ZP3 (rhZP3), mouse ZP and two MTx channel blockers (U73122 and U73343), we investigated and compared the MTx- and ZP-induced ARs in human and mouse spermatozoa. Herein, we report that MTx induced AR and elevated intracellular Ca²(+) ([Ca²(+)](i)) in human spermatozoa, both of which were blocked by U73122 and U73343. These two compounds also inhibited the MTx-induced AR in mouse spermatozoa. In disagreement with our previous proposal, the AR triggered by rhZP3 or mouse ZP was not blocked by U73343, indicating that in human and mouse spermatozoa, the AR induction by the physiological ligands or by MTx occurred through distinct pathways. U73122, but not U73343 (inactive analogue), can block phospholipase C (PLC). Another PLC inhibitor, edelfosine, also blocked the rhZP3- and ZP-induced ARs. These findings confirmed the participation of a PLC-dependent signalling pathway in human and mouse zona protein-induced AR. Notably, edelfosine also inhibited the MTx-induced mouse sperm AR but not that of the human, suggesting that toxin-induced AR is PLC-dependent in mice and PLC-independent in humans.


Subject(s)
Acrosome Reaction/physiology , Calcium Channels/metabolism , Marine Toxins/pharmacology , Oxocins/pharmacology , Spermatozoa/metabolism , Zona Pellucida/metabolism , Acrosome Reaction/drug effects , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Estrenes/pharmacology , Humans , Male , Mice , Pyrrolidinones/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Sperm Capacitation/physiology , Spermatozoa/drug effects , Type C Phospholipases/antagonists & inhibitors , Zona Pellucida/drug effects
11.
J Biol Chem ; 285(21): 16302-14, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20236935

ABSTRACT

Regulated secretion is a central issue for the specific function of many cells; for instance, mammalian sperm acrosomal exocytosis is essential for egg fertilization. Sphingosine 1-phosphate is a bioactive sphingolipid that regulates crucial physiological processes. Here we report that this lipid triggers acrosomal exocytosis in human sperm by a mechanism involving a G(i)-coupled receptor. Real-time imaging showed a remarkable increase of cytosolic calcium upon activation with sphingosine 1-phosphate and pharmacological experiments indicate that the process requires extracellular calcium influx through voltage and store-operated calcium channels and efflux from intracellular stores through inositol 1,4,5-trisphosphate-sensitive calcium channels. Sphingosine 1-phosphate-induced exocytosis requires phospholipase C and protein kinase C activation. We investigated possible sources of the lipid. Western blot indicates that sphingosine kinase 1 is present in spermatozoa. Indirect immunofluorescence showed that phorbol ester, a potent protein kinase C activator that can also trigger acrosomal exocytosis, redistributes sphingosine kinase 1 to the acrosomal region. Functional assays showed that phorbol ester-induced exocytosis depends on the activation of sphingosine kinase 1. Furthermore, incorporation of (32)P to sphingosine demonstrates that cells treated with the phorbol ester increase their sphingosine kinase activity that yields sphingosine 1-phosphate. We present here the first evidence indicating that human spermatozoa produce sphingosine 1-phosphate when challenged with an exocytic stimulus. These observations point to a new role of sphingosine 1-phosphate in a signaling cascade that facilitates acrosome reaction providing some clues about novel lipid molecules involved in exocytosis.


Subject(s)
Acrosome/metabolism , Exocytosis/physiology , Lysophospholipids/metabolism , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Enzyme Activation/drug effects , Enzyme Activation/physiology , Exocytosis/drug effects , Female , Humans , Male , Phorbol Esters/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction/drug effects , Sphingosine/metabolism
12.
PLoS One ; 4(6): e6095, 2009 Jun 30.
Article in English | MEDLINE | ID: mdl-19582168

ABSTRACT

BACKGROUND: The transient receptor potential channel (TRP) family includes more than 30 proteins; they participate in various Ca(2+) dependent processes. TRPs are functionally diverse involving thermal, chemical and mechanical transducers which modulate the concentration of intracellular Ca(2+) ([Ca(2+)]i). Ca(2+) triggers and/or regulates principal sperm functions during fertilization such as motility, capacitation and the acrosome reaction. Nevertheless, the presence of the TRPM subfamily in sperm has not been explored. PRINCIPAL FINDINGS: Here we document with RT-PCR, western blot and immunocitochemistry analysis the presence of TRPM8 in human sperm. We also examined the participation of this channel in sperm function using specific agonists (menthol and temperature) and antagonists (BCTC and capsazepine). Computer-aided sperm analysis revealed that menthol did not significantly alter human sperm motility. In contrast, menthol induced the acrosome reaction in human sperm. This induction was inhibited about 70% by capsazepine (20 microM) and 80% by BCTC (1.6 microM). Activation of TRPM8 either by temperature or menthol induced [Ca(2+)]i increases in human sperm measured by fluorescence in populations or individual sperm cells, effect that was also inhibited by capsazepine (20 microM) and BCTC (1.6 microM). However, the progesterone and ZP3-induced acrosome reaction was not inhibited by capsazepine or BCTC, suggesting that TRPM8 activation triggers this process by a different signaling pathway. CONCLUSIONS: This is the first report dealing with the presence of a thermo sensitive channel (TRPM8) in human sperm. This channel could be involved in cell signaling events such as thermotaxis or chemotaxis.


Subject(s)
Spermatozoa/metabolism , TRPM Cation Channels/physiology , Acrosome Reaction , Calcium/metabolism , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Chemotaxis , Fluorescent Antibody Technique, Indirect , Humans , Male , Models, Biological , Progesterone/metabolism , Pyrazines/pharmacology , Pyridines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sperm Motility , TRPM Cation Channels/metabolism
13.
J Biol Chem ; 284(37): 24825-39, 2009 Sep 11.
Article in English | MEDLINE | ID: mdl-19546222

ABSTRACT

Exocytosis of the acrosome (the acrosome reaction) relies on cAMP production, assembly of a proteinaceous fusion machinery, calcium influx from the extracellular medium, and mobilization from inositol 1,4,5-trisphosphate-sensitive intracellular stores. Addition of cAMP to human sperm suspensions bypasses some of these requirements and elicits exocytosis in a protein kinase A- and extracellular calcium-independent manner. The relevant cAMP target is Epac, a guanine nucleotide exchange factor for the small GTPase Rap. We show here that a soluble adenylyl cyclase synthesizes the cAMP required for the acrosome reaction. Epac stimulates the exchange of GDP for GTP on Rap1, upstream of a phospholipase C. The Epac-selective cAMP analogue 8-pCPT-2'-O-Me-cAMP induces a phospholipase C-dependent calcium mobilization in human sperm suspensions. In addition, our studies identify a novel connection between cAMP and Rab3A, a secretory granule-associated protein, revealing that the latter functions downstream of soluble adenylyl cyclase/cAMP/Epac but not of Rap1. Challenging sperm with calcium or 8-pCPT-2'-O-Me-cAMP boosts the exchange of GDP for GTP on Rab3A. Recombinant Epac does not release GDP from Rab3A in vitro, suggesting that the Rab3A-GEF activation by cAMP/Epac in vivo is indirect. We propose that Epac sits at a critical point during the exocytotic cascade after which the pathway splits into two limbs, one that assembles the fusion machinery into place and another that elicits intracellular calcium release.


Subject(s)
Apolipoproteins A/metabolism , Exocytosis/physiology , GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Telomere-Binding Proteins/metabolism , Acrosome Reaction , Apolipoprotein A-V , Calcium/metabolism , Fertilization , Guanosine Triphosphate/metabolism , Humans , Male , Models, Biological , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Shelterin Complex , Spermatozoa/metabolism
14.
PLoS One ; 4(3): e4963, 2009.
Article in English | MEDLINE | ID: mdl-19305511

ABSTRACT

Hydrocephalus with hop gait (hyh) is a recessive inheritable disease that arose spontaneously in a mouse strain. A missense mutation in the Napa gene that results in the substitution of a methionine for isoleucine at position 105 (M105I) of alphaSNAP has been detected in these animals. alphaSNAP is a ubiquitous protein that plays a key role in membrane fusion and exocytosis. In this study, we found that male hyh mice with a mild phenotype produced morphologically normal and motile sperm, but had a strongly reduced fertility. When stimulated with progesterone or A23187 (a calcium ionophore), sperm from these animals had a defective acrosome reaction. It has been reported that the M105I mutation affects the expression but not the function of the protein. Consistent with an hypomorphic phenotype, the testes and epididymides of hyh mice had low amounts of the mutated protein. In contrast, sperm had alphaSNAP levels indistinguishable from those found in wild type cells, suggesting that the mutated protein is not fully functional for acrosomal exocytosis. Corroborating this possibility, addition of recombinant wild type alphaSNAP rescued exocytosis in streptolysin O-permeabilized sperm, while the mutant protein was ineffective. Moreover, addition of recombinant alphaSNAP. M105I inhibited acrosomal exocytosis in permeabilized human and wild type mouse sperm. We conclude that the M105I mutation affects the expression and also the function of alphaSNAP, and that a fully functional alphaSNAP is necessary for acrosomal exocytosis, a key event in fertilization.


Subject(s)
Acrosome Reaction/physiology , Mice, Mutant Strains , Point Mutation , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/genetics , Animals , Epididymis/metabolism , Exocytosis/physiology , Female , Fertility/physiology , Fertilization in Vitro , Humans , Male , Mice , Mice, Inbred C57BL , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Spermatozoa/cytology , Spermatozoa/metabolism , Testis/cytology , Testis/metabolism
15.
FASEB J ; 21(14): 4121-30, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17625073

ABSTRACT

The acrosome reaction is a regulated Ca2+-dependent secretion event required for sperm-egg interaction. Previous studies indicate that the process requires Rab3-dependent tethering of membranes, SNARE complex assembly, and Ca2+-mediated activation of synaptotagmin. Sperm are transcriptionally and translationally inactive; hence, most studies of the exocytosis mechanism are limited to membrane-permeant reagents. The effect of proteins involved in exocytosis has been assessed only in permeabilized cells. Polyarginine peptides are a powerful tool for delivering macromolecules to cells. Most reports indicate that membrane translocation of arginine-containing proteins requires endocytosis; therefore, this strategy might not be useful in sperm. However, our results indicate that GST and Rab3A, when fused with an arginine-rich peptide, were able to translocate into sperm. Moreover, membrane-permeant Rab3A initiated exocytosis when prenylated and activated with GTP. We show here that a key event after the cytoplasmic Ca2+ increase caused by progesterone is the activation of Rab3A. When active Rab3A is introduced into sperm, Ca2+ in the extracellular medium and in the cytoplasm is dispensable. However, a Ca2+ efflux from inside the acrosome is still required to achieve exocytosis. In conclusion, arginine-containing proteins can penetrate the sperm plasma membrane and thus are valuable tools to study sperm physiology in intact cells.


Subject(s)
Acrosome/metabolism , Cell Membrane Permeability/physiology , Exocytosis/physiology , Spermatozoa/metabolism , rab3A GTP-Binding Protein/physiology , Acrosome/enzymology , Animals , CHO Cells , Cricetinae , Cricetulus , Enzyme Activation/physiology , Female , Guanosine Triphosphate/physiology , Humans , Male , Protein Prenylation , Spermatozoa/enzymology
16.
J Biol Chem ; 282(36): 26335-43, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17613520

ABSTRACT

Regulated secretion is a fundamental process underlying the function of many cell types. In particular, acrosomal exocytosis in mammalian sperm is essential for egg fertilization. Regulated secretion requires SNARE proteins and, in neurons, also synaptotagmin I and complexin. Recent reports suggest that complexin imposes a fusion block that is released by Ca(2+) and synaptotagmin I. However, no direct evidence for this model in secreting cells has been provided and whether this complexin/synaptotagmin interplay functions in other types of secretion is unknown. In this report, we show that the C2B domain of synaptotagmin VI and an anti-complexin antibody blocked the formation of trans SNARE complexes in permeabilized human sperm, and that this effect was reversed by adding complexin. In contrast, an excess of complexin stopped exocytosis at a later step, when SNAREs were assembled in loose trans complexes. Interestingly, this blockage was released by the addition of the synaptotagmin VI C2B domain in the presence of Ca(2+). We have previously demonstrated that the activity of this domain is regulated by protein kinase C-mediated phosphorylation. Here, we show that a phosphomimetic mutation in the polybasic region of the C2B domain strongly affects its Ca(2+) and phospholipids binding properties. Importantly, this mutation completely abrogates its ability to rescue the complexin block. Our results show that the functional interplay between complexin and synaptotagmin has a central role in a physiological secretion event, and that this interplay can be modulated by phosphorylation of the C2B domain.


Subject(s)
Acrosome/metabolism , Exocytosis/physiology , Nerve Tissue Proteins/metabolism , Synaptotagmin I/metabolism , Synaptotagmins/metabolism , Adaptor Proteins, Vesicular Transport , Animals , Calcium/metabolism , Exocytosis/drug effects , Fertilization/physiology , Humans , Male , Nerve Tissue Proteins/pharmacology , Phosphorylation/drug effects , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Rats , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Synaptotagmin I/pharmacology , Synaptotagmins/pharmacology
17.
PLoS Biol ; 3(10): e323, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16131227

ABSTRACT

The dynamics of SNARE assembly and disassembly during membrane recognition and fusion is a central issue in intracellular trafficking and regulated secretion. Exocytosis of sperm's single vesicle--the acrosome--is a synchronized, all-or-nothing process that happens only once in the life of the cell and depends on activation of both the GTP-binding protein Rab3 and of neurotoxin-sensitive SNAREs. These characteristics make acrosomal exocytosis a unique mammalian model for the study of the different phases of the membrane fusion cascade. By using a functional assay and immunofluorescence techniques in combination with neurotoxins and a photosensitive Ca2+ chelator we show that, in unactivated sperm, SNAREs are locked in heterotrimeric cis complexes. Upon Ca2+ entry into the cytoplasm, Rab3 is activated and triggers NSF/alpha-SNAP-dependent disassembly of cis SNARE complexes. Monomeric SNAREs in the plasma membrane and the outer acrosomal membrane are then free to reassemble in loose trans complexes that are resistant to NSF/alpha-SNAP and differentially sensitive to cleavage by two vesicle-associated membrane protein (VAMP)-specific neurotoxins. Ca2+ must be released from inside the acrosome to trigger the final steps of membrane fusion that require fully assembled trans SNARE complexes and synaptotagmin. Our results indicate that the unidirectional and sequential disassembly and assembly of SNARE complexes drive acrosomal exocytosis.


Subject(s)
Acrosome/physiology , Calcium/pharmacology , Exocytosis/physiology , SNARE Proteins/metabolism , Spermatozoa/metabolism , rab3A GTP-Binding Protein/physiology , Acrosome Reaction/physiology , Botulinum Toxins/pharmacology , Botulinum Toxins, Type A , Calcimycin/pharmacology , Calcium/metabolism , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Ethylenediamines/pharmacology , Fluorescent Antibody Technique , Humans , Male , Permeability/drug effects , Protein Structure, Quaternary/drug effects , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Spermatozoa/drug effects , Synaptosomal-Associated Protein 25/metabolism , Synaptotagmins/physiology , Tetanus Toxin/pharmacology , Vesicle-Associated Membrane Protein 2/metabolism
18.
Dev Biol ; 285(2): 422-35, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16111671

ABSTRACT

We have previously reported that synaptotagmin VI is present in human sperm cells and that a recombinant protein containing the C2A and C2B domains abrogates acrosomal exocytosis in permeabilized spermatozoa, an effect that was regulated by phosphorylation. In this report, we show that each individual C2 domain blocks acrosomal exocytosis. The inhibitory effect was completely abrogated by phosphorylation of the domains with purified PKCbetaII. We found by site-directed mutagenesis that Thr418 and/or Thr419 in the polybasic region (KKKTTIK) of the C2B domain--a key region for the function of synaptotagmins--are the PKC target that regulates its inhibitory effect on acrosomal exocytosis. Similarly, we showed that Thr284 in the polybasic region of C2A (KCKLQTR) is the target for PKC-mediated phosphorylation in this domain. An antibody that specifically binds to the phosphorylated polybasic region of the C2B domain recognized endogenous phosphorylated synaptotagmin in the sperm acrosomal region. The antibody was inhibitory only at early stages of exocytosis in sperm acrosome reaction assays, and the immunolabeling decreased upon sperm stimulation, indicating that the protein is dephosphorylated during acrosomal exocytosis. Our results indicate that acrosomal exocytosis is regulated through the PKC-mediated phosphorylation of conserved threonines in the polybasic regions of synaptotagmin VI.


Subject(s)
Acrosome/physiology , Exocytosis/physiology , Models, Biological , Protein Kinase C/metabolism , Synaptotagmins/metabolism , Acrosome/metabolism , Amino Acid Sequence , Analysis of Variance , DNA Primers , Fluorescent Antibody Technique, Indirect , Humans , Male , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C beta , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...