Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Mol Hum Reprod ; 20(5): 392-408, 2014 May.
Article in English | MEDLINE | ID: mdl-24442342

ABSTRACT

Whilst fertilizing capacity depends upon a K(+) conductance (GK) that allows the spermatozoon membrane potential (Vm) to be held at a negative value, the characteristics of this conductance in human sperm are virtually unknown. We therefore studied the biophysical/pharmacological properties of the K(+) conductance in spermatozoa from normal donors held under voltage/current clamp in the whole cell recording configuration. Our standard recording conditions were designed to maintain quasi-physiological, Na(+), K(+) and Cl(-) gradients. Experiments that explored the effects of ionic substitution/ion channel blockers upon membrane current/potential showed that resting Vm was dependent upon a hyperpolarizing K(+) current that flowed via channels that displayed only weak voltage dependence and limited (∼7-fold) K(+) versus Na(+) selectivity. This conductance was blocked by quinidine (0.3 mM), bupivacaine (3 mM) and clofilium (50 µM), NNC55-0396 (2 µM) and mibefradil (30 µM), but not by 4-aminopyridine (2 mM, 4-AP). Progesterone had no effect upon the hyperpolarizing K(+) current. Repolarization after a test depolarization consistently evoked a transient inward 'tail current' (ITail) that flowed via a second population of ion channels with poor (∼3-fold) K(+) versus Na(+) selectivity. The activity of these channels was increased by quinidine, 4-AP and progesterone. Vm in human sperm is therefore dependent upon a hyperpolarizing K(+) current that flows via channels that most closely resemble those encoded by Slo3. Although 0.5 µM progesterone had no effect upon these channels, this hormone did activate the pharmacologically distinct channels that mediate ITail. In conclusion, this study reveals three functionally and pharmacologically distinct cation channels: Ik, ITail, ICatSper.


Subject(s)
Calcium Channels/metabolism , Fertilization , Patch-Clamp Techniques , Potassium Channels/metabolism , Sodium Channels/metabolism , Spermatozoa/metabolism , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Fertilization/drug effects , Humans , Hydrogen-Ion Concentration , Male , Membrane Potentials , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Progesterone/pharmacology , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Spermatozoa/drug effects
4.
Soc Reprod Fertil Suppl ; 63: 237-55, 2007.
Article in English | MEDLINE | ID: mdl-17566277

ABSTRACT

Sperm dysfunction is the single most common defined cause of infertility. One in 15 men is sub-fertile and the condition is increasing in frequency. However, the diagnosis is poor and, excluding assisted conception, there is no treatment. The reason for this is our limited understanding of the biochemical, molecular and genetic functions of the spermatozoon. The underlying premise of our research programme is to establish a rudimentary understanding of the processes necessary for successful fertilisation. In this manuscript, we detail advances in our understanding of calcium signalling in the cell and outline genetic and proteomic technologies that are being used to improve the diagnosis of the condition.


Subject(s)
Calcium Signaling/physiology , Infertility, Male/diagnosis , Spermatozoa/physiology , Calcium/metabolism , Humans , Infertility, Male/pathology , Infertility, Male/therapy , Male , Proteomics , Sodium-Calcium Exchanger/metabolism
5.
Dev Biol ; 302(1): 324-32, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17054937

ABSTRACT

Human spermatozoa stimulated with progesterone (a product of the cumulus and thus encountered by sperm prior to fertilization in vivo) apparently mobilize Ca(2+) and respond very differently according to the way in which the steroid is presented. A progesterone concentration ramp (0-3 microM) induces [Ca(2+)](i) oscillations (repetitive store mobilization) which modify flagellar beating, whereas bolus application of micromolar progesterone causes a single large transient (causing acrosome reaction) which is apparently dependent upon Ca(2+) influx. We have investigated Ca(2+)-mobilization and functional responses in human sperm exposed to 3 muM progesterone. The [Ca(2+)](i) response to progesterone was abolished by 4 min incubation in 0 Ca(2+) medium (2 mM EGTA) but in nominally Ca(2+)-free medium (no added Ca(2+); 0 EGTA) a smaller, slow response occurred. Single cell imaging showed a similar effect of nominally Ca(2+)-free medium and approximately 5% of cells generated a small transient even in the presence of EGTA. When cells were exposed to EGTA-containing saline (5 min) and then returned to nominally Ca(2+)-free medium before stimulation, the [Ca(2+)](i) transient was greatly delayed (approximately 50 s) and rise time was doubled in comparison to cells not subjected to EGTA pre-treatment. We conclude that mobilization of stored Ca(2+) contributes a 'slow' component to the progesterone-induced [Ca(2+)](i) transient and that incubation in EGTA-buffered saline is able rapidly to deplete this store. Analysis of flagellar activity induced by 3 muM progesterone showed an effect (modified beating) associated with the [Ca(2+)](i) transient, in >80% of cells bathed in nominally Ca(2+)-free medium. This was reduced greatly in cells subjected to 5 min EGTA pre-treatment. The store-mediated transient showed a pharmacological sensitivity similar to that of progesterone-induced [Ca(2+)](i) oscillations (consistent with filling of the store by an SPCA) suggesting that the transient induced by micromolar progesterone is a 'single shot' activation of the same store that generates Ca(2+) oscillations.


Subject(s)
Calcium Signaling , Calcium/metabolism , Progesterone/metabolism , Spermatozoa/metabolism , Acrosome/metabolism , Acrosome Reaction , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Male , Progesterone/pharmacology , Spermatozoa/cytology , Thapsigargin/pharmacology
6.
Hum Reprod Update ; 12(3): 253-67, 2006.
Article in English | MEDLINE | ID: mdl-16338990

ABSTRACT

Ca(2+) is a ubiquitous intracellular messenger which encodes information by temporal and spatial patterns of concentration. In spermatozoa, several key functions, including acrosome reaction and motility, are regulated by cytoplasmic Ca(2+) concentration. Despite the very small size and apparent structural simplicity of spermatozoa, evidence is accumulating that they possess sophisticated mechanisms for regulation of cytoplasmic Ca(2+) concentration and generation of complex Ca(2+) signals. In this review, we consider the various components of the Ca(2+)-signalling 'toolkit' that have been characterized in somatic cells and summarize the evidence for their presence and activity in spermatozoa. In particular, data accumulated over the last few years show that spermatozoa possess one (and probably two) Ca(2+) stores as well as a range of plasma membrane pumps and channels. Selective regulation of the various components of the 'toolkit' by agonists probably allows spermatozoa to generate localized Ca(2+) signals despite their very small cytoplasmic volume, permitting the discrete and selective activation of cell functions.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Calcium/metabolism , Spermatozoa/metabolism , Calcium/agonists , Calcium-Transporting ATPases/metabolism , Humans , Male
7.
Mol Hum Reprod ; 11(9): 683-91, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16192296

ABSTRACT

We have investigated the reversibility of biochemical and physiological changes that occur upon suspension of ejaculated human spermatozoa during in vitro capacitation. Cells were swum up in a simple HEPES-based saline [lacking bicarbonate and bovine serum albumin (BSA)], then resuspended either in supplemented Earle's balanced salt solution (sEBSS) (25 mM bicarbonate) with 0.3% BSA (for in vitro capacitation) or in medium-lacking bicarbonate and/or BSA. Progesterone-induced acrosome reaction (AR) developed during in vitro capacitation (6 h). A progesterone-induced [Ca2+]i signal was detectable in cells maintained in the simple HEPES-based saline, but upon transfer to sEBSS, the response increased three- to four-fold, saturating within <30 min. Serine/threonine phosphorylation saturated within minutes of resuspension, but tyrosine phosphorylation developed over 3 h. Return of cells to non-capacitating conditions caused reversal of all capacitation-dependent changes. The [Ca2+]i signal reverted to its 'uncapacitated' size within <30 min. Protein phosphorylation reversed gradually and could be reinduced (kinetics resembling the first response) upon resuspension in sEBSS. The ability of cells to undergo progesterone-induced AR fell to levels similar to those in uncapacitated cells within 1 h of resuspension in medium not supporting capacitation. Loss of protein phosphorylation occurred only in the absence of both bicarbonate and BSA, but effects on [Ca2+]i signalling and AR could be seen after removal of only one of these factors. We conclude that key events in the capacitation of human spermatozoa are both reversible and repeatable.


Subject(s)
Acrosome Reaction , Bicarbonates/pharmacology , Serum Albumin, Bovine/pharmacology , Sperm Capacitation , Spermatozoa/drug effects , Calcium Signaling , Cells, Cultured , Culture Media , Humans , Male , Phosphorylation , Progesterone , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Sodium Bicarbonate/pharmacology , Spermatozoa/metabolism
8.
Mol Hum Reprod ; 11(7): 523-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16123082

ABSTRACT

Sperm capacitation is a prerequisite for successful in vitro fertilization (IVF) and therefore a focus of sperm preparation in IVF laboratories. The technology of IVF is, therefore, potentially valuable in advancing our understanding of the molecular processes that occur during sperm capacitation. We have investigated sperm capacitation induced by a commercial IVF medium compared to that occurring in standard capacitating medium (CM) typically used in a nonclinical setting. Percoll-washed spermatozoa were resuspended in Cook Sydney IVF medium, Cook Sydney IVF sperm buffer, Earle's balanced salt medium (capacitating medium) or a modified Earle's balanced salt medium [non-capacitating medium (NCM)] for up to 120 min at 37 degrees C and, if applicable, in the presence of 5% CO2 in air. Sperm protein kinase A (PKA) activity, PKA-dependent serine/threonine phosphorylation, tyrosine phosphorylation, hyperactivation and progesterone-induced acrosome reaction were evaluated. IVF medium was shown to accelerate sperm capacitation (compared with capacitating medium) as determined by tyrosine phosphorylation, sperm hyperactivation and progesterone-induced acrosome reaction. This effect was not associated with enhanced activation of PKA or increased levels of serine/threonine phosphorylation. In contrast, IVF sperm buffer (used for sperm preparation) did not stimulate sperm capacitation when incubated for up to 90 min. We have shown that different capacitating media vary strikingly in their efficacy and that this difference reflects activation of a pathway other than the well-characterized activation of soluble adenylyl cyclase/cAMP/PKA.


Subject(s)
Acrosome Reaction/physiology , Culture Media , Cyclic AMP-Dependent Protein Kinases/metabolism , Fertilization in Vitro , Progesterone/physiology , Spermatozoa/metabolism , Tyrosine/metabolism , Cyclic AMP-Dependent Protein Kinases/physiology , Dimethyl Sulfoxide , Enzyme Activation/physiology , Humans , Male , Phosphorylation , Serine/metabolism , Sperm Capacitation/physiology , Threonine/metabolism
9.
Dev Biol ; 274(2): 308-17, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15385161

ABSTRACT

Although ion channels are known to be pivotal to sperm function, the technical difficulty of applying electrophysiological techniques to spermatozoa has prevented significant progress in this area. This is due to the cell size and angular shape in combination with their motility. Using a refined technique, specifically for patch clamping spermatozoa, we have made recordings from human cells. This technique permitted approaches which enable functional analysis of sperm ion channels, including acquisition of inside-out patches, generation of averaged currents, and observation of the effects of pharmacological manipulation during prolonged recordings. As well as a low conductance (7 pS) channel and a 25-pS channel, the most striking finding was the presence of very high conductance, 4-aminopyridine-sensitive multistate channels resembling the non-selective cation channel of sea urchin and mouse spermatozoa. Application of 2 mM 4-aminopyridine (a dose sufficient to cause channel blockade) caused an instant and dramatic transition of motility in the sperm population increasing hyperactivated motility by more than 10-fold as assessed by computer-assisted semen analysis. Combined application of patch clamp and pharmacological investigation of mature sperm cells and will permit rapid advances in our understanding the role of ion channels in sperm function.


Subject(s)
4-Aminopyridine/metabolism , Ion Channels/metabolism , Patch-Clamp Techniques , Potassium Channel Blockers/metabolism , Spermatozoa/physiology , 4-Aminopyridine/pharmacology , Animals , Humans , Male , Mice , Patch-Clamp Techniques/instrumentation , Potassium Channel Blockers/pharmacology , Progesterone/pharmacology , Sperm Capacitation , Sperm Motility/drug effects , Sperm Motility/physiology , Spermatozoa/cytology , Spermatozoa/drug effects , Tetraethylammonium/pharmacology
10.
Dev Biol ; 259(1): 71-82, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12812789

ABSTRACT

The steroid progesterone, an agonist of acrosome reaction, induces a biphasic [Ca(2+)](i)-signal in human sperm comprising an initial transient [Ca(2+)](i) elevation, and a subsequent ramp or plateau. Nifedipine, an inhibitor of voltage-operated Ca(2+) channels, inhibits progesterone-induced acrosome reaction in human sperm, but fluorimetric studies have detected no effect of this compound on the progesterone-induced [Ca(2+)](i) signal. We have used single-cell imaging to study the effects of nifedipine on [Ca(2+)](i) signalling in human sperm. Analysis of mean responses from large numbers of cells showed that treatment with nifedipine reduced the duration but not the amplitude of the progesterone-induced [Ca(2+)](i) transient. In control cells, the latency of the transient peak (maximum fluorescence) fell within the range of 30-105 s. In the presence of nifedipine, very few cells peaked "late" (>60 s after application of progesterone). Analysis of transient responses in control cells revealed characteristic "early" and "late" responses, most cells showing both "early" and "late" transients, whereas "late" transients were rare and smaller in the presence of nifedipine. Sustained responses showed strong association with late transients, and occurrence and amplitude of sustained responses were significantly reduced in nifedipine pretreated cells. These findings are consistent with the occurrence of a discrete, nifedipine-sensitive component of the progesterone-induced [Ca(2+)](i) transient that peaks 1-2 min after exposure to the hormone and is crucial for the induction of the sustained [Ca(2+)](i) signal.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium/metabolism , Nifedipine/pharmacology , Progesterone/pharmacology , Spermatozoa/drug effects , Acrosome/drug effects , Dose-Response Relationship, Drug , Humans , Male , Spermatozoa/metabolism
11.
Calcif Tissue Int ; 70(3): 194-203, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11907717

ABSTRACT

Cells of mammalian bone express glutamate receptors. Functional N-methyl-D-aspartate (NMDA) receptors have been demonstrated in human, osteoblastic MG-63 cells, but currents in these cells, unlike those of mammalian neurons, are blocked by Mg(2+) in a voltage-insensitive manner. Differences between the characteristics of NMDA currents in bone cells and in neurons may reflect molecular variation of the receptors or associated molecules, with implications for the role(s) of glutamate in these different tissues and for targeting of ligands/antagonists. To determine whether NMDA receptors in primary bone cells are functional, and whether the currents carried by these receptors resemble those of MG-63 cells or those of mammalian neurons, we have applied the whole cell patch clamp technique to primary cultures of rat osteoblasts. In 0-Mg(2+) saline, 25% of cells showed a slowly developing inward current in response to bath perfusion with 1 mM or 100 microM NMDA. Antibodies against NMDA receptors stained approximately 26% of cells. When NMDA was applied by rapid superfusion, kinetics of the currents were similar to those of neuronal NMDA currents, reaching a peak within 20-30 ms. 1 mM Mg(2+) reduced current amplitude at negative holding potentials and caused the I-V relationship of the currents to adopt a 'J' shape rather than the linear relationship seen in the absence of added Mg(2+). Co-application of glycine (20 microM) with NMDA increased current amplitude by only 18%, suggesting that glycine is released from cells within the cultures. Currents were blocked by (+)-MK-801 and DL-2-amino-5-phosphonovaleric acid. Fluorimetric monitoring of [Ca(2+)](i) using fura-2 showed that, in Mg(2+)-free medium, NMDA caused a sustained rise in [Ca(2+)](i) that could be reversed by subsequent application of MK-801. We conclude that rat femoral osteoblasts express functional NMDA receptors and that these receptors differ from those previously identified in MG-63 cells. NMDA receptors of primary osteoblasts show a 'classical' voltage-sensitive Mg(2+) block, similar to that seen in neuronal NMDA receptors, and will therefore function as detectors of coincident receptor activation and membrane depolarization.


Subject(s)
Calcium/metabolism , Magnesium/pharmacology , Neurons/physiology , Osteoblasts/physiology , Receptors, N-Methyl-D-Aspartate/physiology , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Dizocilpine Maleate/pharmacology , Electrophysiology , Excitatory Amino Acid Antagonists/pharmacology , Femur/cytology , Femur/physiology , Glycine/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , N-Methylaspartate/pharmacology , Neurons/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Patch-Clamp Techniques , Perfusion , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/biosynthesis
12.
Mol Hum Reprod ; 8(4): 326-32, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11912280

ABSTRACT

Previous studies have implicated receptor tyrosine kinases in progesterone-induced [Ca2+]i signalling, and consequent induction of the acrosome reaction, in human spermatozoa. We have investigated the effects of tyrosine kinase inhibition on [Ca2+]i responses in large numbers of individual human spermatozoa. Genistein (5, 50 and 250 micromol/l), an inhibitor of receptor-linked tyrosine kinases, significantly inhibited the progesterone-induced acrosome reaction (P < 0.05). However, we could detect no effect of genistein on progesterone-induced [Ca2+]i signalling. In control experiments, application of progesterone induced a significant transient [Ca2+]i response in approximately 77% of cells and a sustained [Ca2+]i ramp/plateau in approximately 48% of cells (n = 26; 5411 cells). In preparations pretreated with genistein (50 micromol/l), significant transient and sustained responses were detected in 69.5 and 39.1% of cells respectively (n = 5; 1109 cells). The amplitudes of both transient and sustained [Ca2+]i responses were similar in control and genistein-pretreated preparations. Tyrphostin A47 (100 micromol/l), another receptor tyrosine kinase inhibitor, also failed to inhibit either the transient or sustained [Ca2+]i response (n = 3; 468 cells). Assessment of tyrosine phosphorylation of two sperm proteins (p105/81) showed greatly increased levels of phosphotyrosine in response to capacitation but a negligible increase in response to progesterone stimulation. Pretreatment with genistein (50 and 250 micromol/l) decreased capacitation-induced tyrosine phosphorylation and resulted in a loss of phosphorylation in response to progesterone treatment. We conclude that neither the transient nor sustained phases of the progesterone-induced [Ca2+]i response require receptor tyrosine kinase signalling. Previous reports of modulation of the progesterone-induced [Ca2+]i signal by tyrosine kinase inhibition probably reflect inhibition of the acrosome reaction.


Subject(s)
Calcium Signaling/drug effects , Enzyme Inhibitors/pharmacology , Progesterone/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Spermatozoa/metabolism , Acrosome/enzymology , Acrosome/metabolism , Blotting, Western , Calcium/metabolism , Genistein/pharmacology , Humans , In Vitro Techniques , Male , Progesterone/metabolism , Signal Transduction/physiology , Spermatozoa/enzymology , Tyrosine/metabolism , Tyrphostins/pharmacology
14.
Biochem Biophys Res Commun ; 282(2): 536-42, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11401493

ABSTRACT

Voltage operated calcium channels (VOCCs) are important in stimulus-response coupling in osteoblasts. We have investigated the expression of VOCCs in the mouse osteocyte cell line, MLO-Y4. Using the whole-cell patch clamp technique we were unable to detect any VOCC currents (n = 436) even in the presence of the L-type VOCC agonist Bay K 8644 (n = 350). Reverse transcription polymerase chain reaction (RT-PCR), using primers to detect alpha(1C), alpha(1D), and alpha(1G) VOCC subunits (all of which are expressed in primary osteoblasts), did not generate detectable products with mRNA from MLO-Y4 cells. However, after treatment with physiological levels of hormones, VOCC alpha(1) subunit mRNAs were detected in MLO-Y4 cells. PTH, 17beta-estradiol, and dexamethasone-treatment induced expression of L-type (alpha(1C), alpha(1D)) subunit transcripts. ATP-treatment induced expression of T-type (alpha(1G)) transcripts. Using whole-cell patch clamp we detected VOCC currents in 5-10% of cells after treatment. Current characteristics (L- or T-type) were consistent with the transcript expressed.


Subject(s)
Calcium Channels/metabolism , Hormones/pharmacology , Osteocytes/drug effects , Osteocytes/metabolism , Adenosine Triphosphate/pharmacology , Animals , Base Sequence , Calcium Channels/chemistry , Calcium Channels/genetics , Cell Line , DNA Primers/genetics , Dexamethasone/pharmacology , Estradiol/pharmacology , Gene Expression Regulation/drug effects , Mice , Parathyroid Hormone/pharmacology , Patch-Clamp Techniques , Protein Subunits , Reverse Transcriptase Polymerase Chain Reaction
15.
Bone ; 28(1): 29-37, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11165940

ABSTRACT

Osteocytes play an important role in signaling within bone. Communication of osteocytes with each other and with bone lining cells may have a function in mineral homeostasis and mechanotransduction. However, very little is known of the expression of ion channels in these cells. Using the whole-cell patch-clamp technique, we have detected three types of K(+) currents in the mouse osteocyte-like cell line MLO-Y4. The most commonly observed current (48% of cells) activated rapidly (20 msec) in response to depolarizing steps from -40 mV and exhibited voltage-dependent inactivation. The current was inhibited by 20 mmol/L tetraethyl ammonium (TEA) and abolished by intracellular 2 mmol/L 4-aminopyridine (4-AP). Biophysical and pharmacological characteristics of the current differed from those of inactivating K(+) currents in osteoblastic cells. In 22% of cells, a slowly activating, voltage-activated current was observed (threshold at 20-30 mV). This current was TEA insensitive, was abolished by intracellular application of 2 mmol/L 4-AP, and was strongly inhibited by apamin, a selective inhibitor of small conductance (SK) Ca(2+)-activated K(+) channels. A third current developed during whole-cell dialysis (37% of cells). This current showed little voltage sensitivity. It was abolished by intracellular application of 2 mmol/L 4-AP, high-extracellular Ba(2+) (108 mmol/L), or by inclusion of ATP in the intracellular solution, but was insensitive to TEA, apamin, Cs(+), and glibenclamide. None of these currents was affected by replacement of chloride with acetate in the bath or pipette salines. Reverse-transcription polymerase chain reaction confirmed the presence of mRNA for the types 1 and 2 SK channels, but message for the large conductance (BK) Ca(2+)-activated K(+) channel was not detected in these cells. Message for the sulphonylurea receptor SUR2, a subunit of glibenclamide-insensitive ATP-dependent K(+) channels (K(ATP)), was also detected, but the glibenclamide-sensitive SUR1 subunit was not. These data are the first descriptions of SK- and ATP-sensitive, glibenclamide-insensitive channels in cells of osteoblastic lineage. Our findings are consistent with a change in K(+) channel expression during differentiation from osteoblast to osteocyte. K(+) channels of osteocytes will contribute to maintenance of the cell membrane potential and thus may participate in mechanosensitivity and osteocyte intercellular communication. In addition, they may be involved in homeostatic maintenance of the extracellular fluid occupying the periosteocytic space.


Subject(s)
Osteocytes/cytology , Osteocytes/physiology , Potassium Channels/metabolism , 4-Aminopyridine/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Apamin/pharmacology , Cell Differentiation/physiology , Cell Line , Cell Lineage/physiology , Cesium/pharmacokinetics , Chlorides/pharmacology , Collagen , DNA Primers , Gene Expression/physiology , Glyburide/pharmacology , Guanosine Triphosphate/pharmacology , Hypoglycemic Agents/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Patch-Clamp Techniques , Potassium Channels/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sodium Chloride/pharmacology , Tetraethylammonium/pharmacology
16.
J Cell Biochem ; 79(4): 648-61, 2000 Sep 14.
Article in English | MEDLINE | ID: mdl-10996855

ABSTRACT

Femur-derived osteoblasts cultured from rat femora were loaded with Fluo-3 using the AM ester. A quantifiable stretch was applied and [Ca(2+)]i levels monitored by analysis of fluorescent images obtained using an inverted microscope and laser scanning confocal imaging system. Application of a single pulse of tensile strain via an expandable membrane resulted in immediate increase in [Ca(2+)]i in a proportion of the cells, followed by a slow and steady decrease to prestimulation levels. Application of parathyroid hormone (10(-6) M) prior to mechanical stimulation potentiated the load-induced elevation of [Ca(2+)]i. Mechanically stimulating osteoblasts in Ca(2+)-free media or in the presence of either nifedipine (10 microM; L-type Ca(2+)-channel blocker) or thapsigargin (1 microM; depletes intracellular Ca(2+) stores) reduced strain-induced increases in [Ca(2+) ]i. Furthermore, strain-induced increases in [Ca(2+)]i were enhanced in the presence of Bayer K 8644 (500 nm), an agonist of L-type calcium channels. The effects of mechanical strain with and without inhibitors and agonists are described on the total cell population and on single cell responses. Application of strain and strain in the presence of the calcium-channel agonist Bay K 8644 to periosteal-derived osteoblasts increased levels of the extracellular matrix proteins osteopontin and osteocalcin within 24 h postload. This mechanically induced increase in osteopontin and osteocalcin was inhibited by the addition of the calcium-channel antagonist, nifedipine. Our results suggest an important role for L-type calcium channels and a thapsigargin-sensitive component in early mechanical strain transduction pathways in osteoblasts.


Subject(s)
Bone Matrix/metabolism , Calcium Channels/metabolism , Osteoblasts/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Osteoblasts/drug effects , Osteocalcin/biosynthesis , Osteopontin , Parathyroid Hormone/pharmacology , Rats , Sialoglycoproteins/biosynthesis , Stress, Mechanical , Thapsigargin/pharmacology
17.
J Biol Chem ; 275(44): 34252-9, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10950953

ABSTRACT

Osteoblasts and osteoclasts express functional N-methyl-d-aspartate (NMDA) receptors, which participate in regulation of bone matrix. In rat femoral osteoblasts held in whole cell clamp there is a robust NMDA current but little if any response to l-glutamate. We have investigated expression of metabotropic glutamate receptors (mGluRs) in these cells. By reverse transcription polymerase chain reaction, we have detected expression of mGluR1b (but not mGluR1a, 2, 3, 4, 5, or 6). Blockade of mGluRs with (+/-)-alpha-methyl-carboxyphenyl-glycine resulted in an enlarged l-glutamate-induced current that resembled the response to NMDA. Conversely, prior stimulation of mGluRs with trans-(+/-)-1-amino-1, 3-cyclopentanedicarboxylic acid (1S,3R-ACPD; mGluR agonist) reduced the NMDA-induced current by 77%. Monitoring of [Ca(2+)](i) showed that NMDA induced a sustained elevation of [Ca(2+)](i), which was dependent upon [Ca(2+)](o). Treatment with 1S,3R-ACPD generated an initial transient that was independent of [Ca(2+)](o), followed by a sustained, [Ca(2+)](o)-dependent phase, a response consistent with phospholipase C-mediated mobilization of stored Ca(2+). Investigations of the interaction between the two receptors confirmed inhibitory modulation of the NMDA receptor-induced rise in [Ca(2+)](i) by mGluRs. Parathyroid hormone, which also activates phospholipase C in osteoblasts, had a similar inhibitory effect on the NMDA receptor-induced [Ca(2+)](i) response. Elevation of [Ca(2+)](i) mediated by mGluR activation was reduced by subsequent stimulation of NMDA receptors. This is the first description of mGluRs in bone and shows that complex glutamatergic signaling can occur in this tissue.


Subject(s)
Cycloleucine/analogs & derivatives , Osteoblasts/metabolism , Receptor Cross-Talk , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Base Sequence , Calcium/metabolism , Cells, Cultured , Cycloleucine/pharmacology , DNA Primers , Membrane Potentials , Osteoblasts/cytology , Osteoblasts/physiology , Rats , Receptors, Parathyroid Hormone/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
Dev Biol ; 222(2): 326-35, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10837122

ABSTRACT

Fluorimetric studies on progesterone-induced [Ca(2+)](i) signalling in mammalian spermatozoa show both the well-characterised [Ca(2+)](i) transient and a subsequent sustained phase. However, the sustained phase is thought to reflect release of the fluorochrome during the acrosome reaction and has not been subject to critical investigation. We have used single-cell imaging of [Ca(2+)](i) to analyse the progesterone-induced [Ca(2+)](i) response in large numbers (>2000) of capacitated, human spermatozoa. In 70% of cells, treatment with progesterone induced a transient increase, which typically peaked within 1 min and decayed with a similar time course. Upon rapid application of progesterone this response peaked within 5-20 s. In 35% of progesterone-treated spermatozoa a sustained elevation of [Ca(2+)](i) occurred, which became discernible during the falling phase of the transient response and persisted for at least 20 min. Both [Ca(2+)](i) responses were localised to the postacrosomal region. Averaging of large numbers of single cell responses generated traces similar to those seen in fluorimetric studies. Although the sustained response was strongly associated with the initial, transient response, a few spermatozoa generated sustained responses that were not preceded by a significant transient response (5% of cells). It is concluded that a genuine biphasic [Ca(2+)](i) signal is activated by progesterone and that the sustained response is a discrete signalling event with biological significance.


Subject(s)
Acrosome Reaction/drug effects , Calcium Signaling/physiology , Calcium/metabolism , Progesterone/pharmacology , Sperm Capacitation/drug effects , Spermatozoa/physiology , Activity Cycles , Calcium Signaling/drug effects , Humans , Kinetics , Male , Microscopy, Confocal , Reproducibility of Results , Sperm Capacitation/physiology , Spermatozoa/cytology , Spermatozoa/drug effects , Time Factors
19.
J Cell Physiol ; 183(2): 163-71, 2000 May.
Article in English | MEDLINE | ID: mdl-10737892

ABSTRACT

The effects of hormonal modulators of osteoblast function, parathyroid hormone, 1,25(OH)(2)D(3) and prostaglandins on [Ca(2+)](i) in periosteal-derived osteoblasts from rat femurs have been investigated. Our results show that application of parathyroid hormone PTH (10(-5) M) and prostaglandin E(2) (PGE(2)) (4 microM) result in a rapid heterogeneous elevation in [Ca(2+)](i) that, in the case of PTH, is dependent on both extracellular and intracellular sources of calcium. Variable responses to treatments have been found within populations of cells. The PGE(2) response is dose dependent. Treatment with 1,25(OH)(2)D(3) (10(-8) M) induces a brief (60-90 sec) elevation in [Ca(2+)](i) that is almost totally abolished in EGTA-buffered Ca(2+)-free medium. Interactive effects of multiple hormone treatments have been observed. Pretreatment with 1,25(OH)(2)D(3) results in near-total inhibition of the PTH and PGE(2) responses. In conclusion, modulation of [Ca(2+)](i) appears to play a role not only in the direct effects of osteotropic hormones on osteoblasts but also in the synergistic and antagonistic effects between circulating hormones.


Subject(s)
Calcitriol/pharmacology , Calcium/metabolism , Dinoprostone/pharmacology , Osteoblasts/drug effects , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , Aniline Compounds , Animals , Bone Resorption/etiology , Cells, Cultured , Fluorescent Dyes , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Periosteum/cytology , Rats , Second Messenger Systems , Xanthenes
20.
Pflugers Arch ; 438(4): 553-60, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10519151

ABSTRACT

Voltage operated calcium channels (VOCCs) are implicated in osteoblastic mechano- and hormonal transduction. Very little, however, is known about the expression of VOCCs in osteoblasts of load-bearing bones. Here we describe two types of whole-cell calcium current in rat femoral explant-derived osteoblasts. The first is high-voltage activated and sensitive to nifedipine, Bay K8644 and FPL 64176. The second is low-voltage activated and is sensitive to micromolar concentrations of Ni2+. The properties of these two currents are consistent with those of L-type and T-type calcium currents respectively. T-type currents were detected in most cells on the day of passage, the level of expression being significantly lower on subsequent days. L-type currents were also most common on the day of passage but were detected consistently throughout the 4-day period of study. The reverse transcription polymerase chain reaction with non-specific primers directed against all L-type VOCC alpha 1 subunits and then with specific primers directed against sequences from rat brain alpha 1C (L-type), alpha 1D (L-type) and alpha 1G (T-type) VOCC subunits detected transcripts of appropriate size in all four cases. Products from the three sets of specific primer pairs (alpha 1C, alpha 1D, alpha 1G) were sequenced and were identical to their respective rat brain templates.


Subject(s)
Bone and Bones/physiology , Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Osteoblasts/metabolism , Weight-Bearing/physiology , Animals , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/physiology , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/physiology , Cells, Cultured , Electric Conductivity , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...