Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Sci Rep ; 8(1): 2221, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29396456

ABSTRACT

Fluorescent nanodiamonds (FND) are carbon-based nanomaterials that can efficiently incorporate optically active photoluminescent centers such as the nitrogen-vacancy complex, thus making them promising candidates as optical biolabels and drug-delivery agents. FNDs exhibit bright fluorescence without photobleaching combined with high uptake rate and low cytotoxicity. Focusing on FNDs interference with neuronal function, here we examined their effect on cultured hippocampal neurons, monitoring the whole network development as well as the electrophysiological properties of single neurons. We observed that FNDs drastically decreased the frequency of inhibitory (from 1.81 Hz to 0.86 Hz) and excitatory (from 1.61 to 0.68 Hz) miniature postsynaptic currents, and consistently reduced action potential (AP) firing frequency (by 36%), as measured by microelectrode arrays. On the contrary, bursts synchronization was preserved, as well as the amplitude of spontaneous inhibitory and excitatory events. Current-clamp recordings revealed that the ratio of neurons responding with AP trains of high-frequency (fast-spiking) versus neurons responding with trains of low-frequency (slow-spiking) was unaltered, suggesting that FNDs exerted a comparable action on neuronal subpopulations. At the single cell level, rapid onset of the somatic AP ("kink") was drastically reduced in FND-treated neurons, suggesting a reduced contribution of axonal and dendritic components while preserving neuronal excitability.


Subject(s)
Action Potentials/drug effects , Hippocampus/drug effects , Nanodiamonds , Nerve Net/drug effects , Neurons/drug effects , Animals , Cells, Cultured , Hippocampus/physiology , Mice , Models, Biological , Nerve Net/physiology , Neurons/physiology
2.
Cell Calcium ; 51(3-4): 321-30, 2012.
Article in English | MEDLINE | ID: mdl-22317919

ABSTRACT

Voltage gated Ca(2+) channels are effective voltage sensors of plasma membrane which convert cell depolarizations into Ca(2+) signaling. The chromaffin cells of the adrenal medulla utilize a large number of Ca(2+) channel types to drive the Ca(2+)-dependent release of catecholamines into blood circulation, during normal or stress-induced conditions. Some of the Ca(2+) channels expressed in chromaffin cells (L, N, P/Q, R and T), however, do not control only vesicle fusion and catecholamine release. They also subserve a variety of key activities which are vital for the physiological and pathological functioning of the cell, like: (i) shaping the action potentials of electrical oscillations driven either spontaneously or by ACh stimulation, (ii) controlling the action potential frequency of tonic or bursts firing, (iii) regulating the compensatory and excess endocytosis following robust exocytosis and (iv) driving the remodeling of Ca(2+) signaling which occurs during stressors stimulation. Here, we will briefly review the well-established properties of voltage-gated Ca(2+) channels accumulated over the past three decades focusing on the most recent discoveries on the role that L- (Cav1.2, Cav1.3) and T-type (Cav3.2) channels play in the control of excitability, exocytosis and endocytosis of chromaffin cells in normal and stress-mimicking conditions.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Chromaffin Cells/physiology , Action Potentials , Animals , Calcium Signaling , Catecholamines/metabolism , Endocytosis , Exocytosis , Humans , Receptor Cross-Talk
3.
Biosens Bioelectron ; 26(1): 92-8, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20570501

ABSTRACT

The quantal release of oxidizable molecules can be successfully monitored by means of polarized carbon fiber microelectrodes (CFEs) positioned in close proximity to the cell membrane. To partially overcome certain CFE limitations, mainly related to their low spatial resolution and lack of optical transparency, we developed a planar boron-doped nanocrystalline diamond (NCD) prototype, grown on a transparent sapphire wafer. Responsiveness to applied catecholamines as well as the electrochemical and optical properties of the NCD-based device were first characterized by cyclic voltammetry and optical transmittance measurements. By stimulating chromaffin cells positioned on the device with external KCl, well-resolved quantal exocytotic events could be detected either from one NCD microelectrode, or simultaneously from an array of four microelectrodes, indicating that the chip is able to monitor secretory events (amperometric spikes) from a number of isolated chromaffin cells. Spikes detected by the planar NCD device had comparable amplitudes, kinetics and vesicle diameter distributions as those measured by conventional CFEs from the same chromaffin cell.


Subject(s)
Aluminum Oxide/chemistry , Biosensing Techniques/instrumentation , Chromaffin Cells/metabolism , Diamond/chemistry , Microarray Analysis/instrumentation , Microelectrodes , Nanostructures/chemistry , Animals , Cells, Cultured , Conductometry/instrumentation , Crystallization/methods , Equipment Design , Equipment Failure Analysis , Mice , Microchemistry/instrumentation , Nanostructures/ultrastructure , Nanotechnology/instrumentation
4.
Pflugers Arch ; 457(5): 1093-110, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18779976

ABSTRACT

We studied the effects of the cAMP-hydrolyzing enzyme phosphodiesterase type-4 (PDE4) on the L-type Ca(2+) channels (LTCCs) and Ca(2+)-dependent secretion in mouse chromaffin cells (MCCs). The selective PDE4 inhibitor rolipram (3 microM) had a specific potentiating action on Ca(2+) currents of MCCs (40% increase within 3 min). A similar effect was produced by the selective beta(1)-AR agonist denopamine (1 microM) and by the unselective PDEs inhibitor IBMX (100 microM). Rolipram and denopamine actions were selective for LTCCs, and the Ca(2+) current increase remained unchanged if the two compounds were applied simultaneously. This suggests that at rest, LTCCs in MCCs are down-regulated by the low levels of cAMP determined by PDE4 activity and that LTCCs can be up-regulated by either inhibiting PDE4 or activating beta(1)-AR. No other PDEs are likely involved in this specific action. PDE4 inhibition had also a marked effect on the spontaneous firing of resting MCCs and catecholamine secretion. Rolipram up-regulated the LTCCs contributing to the "pace-maker" current underlying action potential (AP) discharges and accelerated the firing rate, with no significant effects on AP waveform. Acceleration of AP firing was also induced by the LTCC-agonist Bay K (1 microM), while nifedipine (3 microM) reduced the firing frequency, suggesting that LTCCs and intracellular cAMP play a key role in setting the pace-maker current regulating MCCs excitability. Rolipram increased also the size of the ready-releasable pool and the quantal content of secretory vesicles without affecting their probability of release. Thus, rolipram acts on MCCs by up-regulating both exocytosis and AP firings. These two processes are effectively down-regulated by PDE4 at rest and can dramatically increase the quantity of released catecholamines when PDE4 is inhibited and/or cAMP is raised.


Subject(s)
Action Potentials/drug effects , Calcium Channels, L-Type/physiology , Chromaffin Cells/physiology , Exocytosis/drug effects , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Calcium Channels, L-Type/drug effects , Chromaffin Cells/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Ethanolamines/pharmacology , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Rolipram/pharmacology , Secretory Vesicles/drug effects , Up-Regulation
5.
Acta Physiol (Oxf) ; 192(2): 233-46, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18021322

ABSTRACT

Voltage-gated Ca2+ channels (Cav) are highly expressed in the adrenal chromaffin cells of mammalian species. Besides shaping action potential waveforms, they are directly involved in the excitation-secretion coupling underlying catecholamine release and, possibly, control other Ca2+-dependent events that originate near the membrane. These functions are shared by a number of Cav channel types (L, N, P/Q, R and T) which have different structure-function characteristics and whose degree of expression changes remarkably among mammalian species. Understanding precisely the functioning of each voltage-gated Ca2+ channels is a crucial task that helps clarifying the Ca2+-dependent mechanisms controlling exocytosis during physiological and pathological conditions. In this paper, we focus on classical and new roles that L- and T-type channels play in the control of chromaffin cell excitability and neurotransmitter release. Interestingly, L-type channels are shown to be implicated in the spontaneous autorhythmicity of chromaffin cells, while T-type channels, which are absent in adult chromaffin cells, are coupled with secretion and can be recruited following long-term beta-adrenergic stimulation or chronic hypoxia. This suggests that like other cells, adrenal chromaffin cells undergo effective remodelling of membrane ion channels and cell functioning during prolonged stress conditions.


Subject(s)
Calcium Channels, L-Type/physiology , Calcium Channels, T-Type/physiology , Calcium/metabolism , Chromaffin Cells/metabolism , Animals , Calcium Signaling , Catecholamines/metabolism , Humans , Ion Channel Gating , Patch-Clamp Techniques
6.
J Physiol ; 584(Pt 1): 149-65, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17690152

ABSTRACT

alpha(1H) T-type channels recruited by beta(1)-adrenergic stimulation in rat chromaffin cells (RCCs) are coupled to fast exocytosis with the same Ca(2+) dependence of high-threshold Ca(2+) channels. Here we show that RCCs exposed to chronic hypoxia (CH) for 12-18 h in 3% O(2) express comparable densities of functional T-type channels that depolarize the resting cells and contribute to low-voltage exocytosis. Following chronic hypoxia, most RCCs exhibited T-type Ca(2+) channels already available at -50 mV with the same gating, pharmacological and molecular features as the alpha(1H) isoform. Chronic hypoxia had no effects on cell size and high-threshold Ca(2+) current density and was mimicked by overnight incubation with the iron-chelating agent desferrioxamine (DFX), suggesting the involvement of hypoxia-inducible factors (HIFs). T-type channel recruitment occurred independently of PKA activation and the presence of extracellular Ca(2+). Hypoxia-recruited T-type channels were partially open at rest (T-type 'window-current') and contributed to raising the resting potential to more positive values. Their block by 50 microm Ni(2+) caused a 5-8 mV hyperpolarization. The secretory response associated with T-type channels could be detected following mild cell depolarizations, either by capacitance increases induced by step depolarizations or by amperometric current spikes induced by increased [KCl]. In the latter case, exocytotic bursts could be evoked even with 2-4 mm KCl and spike frequency was drastically reduced by 50 microm Ni(2+). Chronic hypoxia did not alter the shape of spikes, suggesting that hypoxia-recruited T-type channels increase the number of secreted vesicles at low voltages, without altering the mechanism of catecholamine release and the quantal content of released molecules.


Subject(s)
Calcium Channels, T-Type/metabolism , Catecholamines/metabolism , Chromaffin Cells/metabolism , Exocytosis/physiology , Hypoxia/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Deferoxamine , Hypoxia-Inducible Factor 1/metabolism , Membrane Potentials/physiology , Potassium Chloride/metabolism , Rats , Rats, Sprague-Dawley , Siderophores , Time Factors , Up-Regulation
7.
Cell Calcium ; 42(4-5): 397-408, 2007.
Article in English | MEDLINE | ID: mdl-17561252

ABSTRACT

Voltage-gated L-type (Cav1.2 and Cav1.3) channels are widely expressed in cardiovascular tissues and represent the critical drug-target for the treatment of several cardiovascular diseases. The two isoforms are also abundantly expressed in neuronal and neuroendocrine tissues. In the brain, Cav1.2 and Cav1.3 channels control synaptic plasticity, somatic activity, neuronal differentiation and brain aging. In neuroendocrine cells, they are involved in the genesis of action potential generation, bursting activity and hormone secretion. Recent studies have shown that Cav1.2 and Cav1.3 are also expressed in chromaffin cells but their functional role has not yet been identified despite that L-type channels possess interesting characteristics, which confer them an important role in the control of catecholamine secretion during action potentials stimulation. In intact rat adrenal glands L-type channels are responsible for adrenaline and noradrenaline release following splanchnic nerve stimulation or nicotinic receptor activation. L-type channels can be either up- or down-modulated by membrane autoreceptors following distinct second messenger pathways. L-type channels are tightly coupled to BK channels and activate at relatively low-voltages. In this way they contribute to the action potential hyperpolarization and to the pace-maker current controlling action potential firings. L-type channels are shown also to regulate the fast secretion of the immediate readily releasable pool of vesicles with the same Ca(2+)-efficiency of other voltage-gated Ca(2+) channels. In mouse adrenal slices, repeated action potential-like stimulations drive L-type channels to a state of enhanced stimulus-secretion efficiency regulated by beta-adrenergic receptors. Here we will review all these novel findings and discuss the possible implication for a specific role of L-type channels in the control of chromaffin cells activity.


Subject(s)
Adrenal Glands/physiology , Calcium Channels, L-Type/physiology , Chromaffin Cells/physiology , Action Potentials , Adrenal Glands/cytology , Animals , Chromaffin Cells/metabolism , Electric Conductivity , Exocytosis , Mice , Rats , Signal Transduction
8.
Eur Biophys J ; 36(7): 753-62, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17340096

ABSTRACT

Expression, spatial distribution and specific roles of different Ca(2+) channels in stimulus-secretion coupling of chromaffin cells are intriguing issues still open to discussion. Most of the evidence supports a role of high-voltage activated (HVA) Ca(2+) channels (L-, N-, P/Q- and R-types) in the control of exocytosis: some suggesting a preferential coupling of specific Ca(2+) channel subunits with the secretory apparatus, others favoring the idea of a contribution to secretion proportional to the expression density and gating properties of Ca(2+) channels. In this work we review recent findings and bring new evidence in favor of the hypothesis that also the LVA (low-voltage-activated, T-type) Ca(2+) channels effectively control fast exocytosis near resting potential in adrenal chromaffin cells of adult rats. T-type channels recruited after long-term treatments with pCPT-cAMP (or chronic hypoxia) are shown to control exocytosis with the same efficacy of L-type channels, which are the dominant Ca(2+) channel types expressed in rodent chromaffin cells. A rigorous comparison of T- and L-type channel properties shows that, although operating at different potentials and with different voltage-sensitivity, the two channels possess otherwise similar Ca(2+)-dependence of exocytosis, size and kinetics of depletion of the immediately releasable pool and mobilize vesicles of the same quantal size. Thus, T- and L-type channels are coupled with the same Ca(2+)-efficiency to the secretory apparatus and deplete the same number of vesicles ready for release. The major difference of the secretory signals controlled by the two channels appear to be the voltage range of operation, suggesting the idea that stressful conditions (hypoxia and persistent beta-adrenergic stimulation) can lower the threshold of cell excitability by recruiting new Ca(2+) channels and activate an additional source of catecholamine secretion.


Subject(s)
Calcium Channels, L-Type/physiology , Calcium Channels, T-Type/physiology , Calcium/physiology , Chromaffin Cells/physiology , Exocytosis/physiology , Ion Channel Gating/physiology , Animals , Calcium Channels, L-Type/drug effects , Calcium Channels, T-Type/drug effects , Cells, Cultured , Chromaffin Cells/drug effects , Cyclic AMP/pharmacology , Exocytosis/drug effects , Hypoxia , Ion Channel Gating/drug effects , Rats
9.
Cell Calcium ; 40(2): 147-54, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16759700

ABSTRACT

Evidence is accumulating on a key role of T-type channels in neurotransmitter release. Recent works have brought undisputable proofs that T-type channels are capable of controlling hormone and neurotransmitters release in association with exocytosis of large dense-core and synaptic vesicles. T-type channel-secretion coupling is not as ubiquitous as that shown for N- and P/Q-type channels in central neurons. In this case, the high-density of Cav2 channel types and co-localization to the release sites ensure high rates of vesicle release and synchronous synaptic responses. Nevertheless, when sufficiently expressed in distal dendrites and neurosecretory cells, T-type channels are able to drive the fast fusion of vesicles ready for release during "low-threshold" Ca2+-entry. T-type channels appear effectively coupled to fast vesicle depletion and may possibly regulate other Ca2+-dependent processes like vesicle recycling and vesicle mobilization from a reserve pool that are important mechanisms controlling synaptic activity during sustained stimulation. Here, we will briefly review the main findings that assign a specific task to T-type channels in fast exocytosis discussing their possible involvement in the control of the Ca2+-dependent processes regulating synaptic activity and vesicular hormone release.


Subject(s)
Calcium Channels, T-Type/metabolism , Exocytosis/physiology , Animals , Endocrine System/cytology , Neurons/cytology , Neurons/metabolism
10.
Pflugers Arch ; 453(3): 373-83, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16758226

ABSTRACT

T-type channels are transient low-voltage-activated (LVA) Ca(2+) channels that control Ca(2+) entry in excitable cells during small depolarizations around resting potential. Studies in the past 20 years focused on the biophysical, physiological, and molecular characterization of T-type channels in most tissues. This led to a well-defined picture of the functional role of LVA channels in controlling low-threshold spikes, oscillatory cell activity, muscle contraction, hormone release, cell growth and differentiation. So far, little attention has been devoted to the role of T-type channels in transmitter release, which mainly involves channel types belonging to the high-voltage-activated (HVA) Ca(2+) channel family. However, evidence is accumulating in favor of a unique participation of T-type channels in fast transmitter release. Clear data are now reported in reciprocal synapses of the retina and olfactory bulb, synaptic contacts between primary afferent and second order nociceptive neurons, rhythmic inhibitory interneurons of invertebrates and clonal cell lines transfected with recombinant alpha(1) channel subunits. T-type channels also regulate the large dense-core vesicle release of neuroendocrine cells where Ca(2+) dependence, rate of vesicle release, and size of readily releasable pool appear comparable to those associated to HVA channels. This suggests that when sufficiently expressed and properly located near the release zones, T-type channels can trigger fast low-threshold secretion. In this study, we will review the main findings that assign a specific task to T-type channels in fast exocytosis, discussing their possible involvement in the control of the Ca(2+)-dependent processes regulating exocytosis like vesicle depletion and vesicle recycling.


Subject(s)
Calcium Channels, T-Type/physiology , Exocytosis/physiology , Animals , Calcium/physiology , Chromaffin Cells/metabolism , Humans , Neurotransmitter Agents/metabolism , Patch-Clamp Techniques , Synaptic Vesicles/physiology
11.
Biophys J ; 90(5): 1830-41, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16361341

ABSTRACT

We have studied the functional role of CaV3 channels in triggering fast exocytosis in rat chromaffin cells (RCCs). CaV3 T-type channels were selectively recruited by chronic exposures to cAMP (3 days) via an exchange protein directly activated by cAMP (Epac)-mediated pathway. Here we show that cAMP-treated cells had increased secretory responses, which could be evoked even at very low depolarizations (-50, -40 mV). Potentiation of exocytosis in cAMP-treated cells did not occur in the presence of 50 microM Ni2+, which selectively blocks T-type currents in RCCs. This suggests that the "low-threshold exocytosis" induced by cAMP is due to increased Ca2+ influx through cAMP-recruited T-type channels, rather than to an enhanced secretion downstream of Ca2+ entry, as previously reported for short-term cAMP treatments (20 min). Newly recruited T-type channels increase the fast secretory response at low voltages without altering the size of the immediately releasable pool. They also preserve the Ca2+ dependence of exocytosis, the initial speed of vesicle depletion, and the mean quantal size of single secretory events. All this indicates that cAMP-recruited CaV3 channels enhance the secretory activity of RCCs at low voltages by coupling to the secretory apparatus with a Ca2+ efficacy similar to that of already existing high-threshold Ca2+ channels. Finally, using RT-PCRs we found that the fast inactivating low-threshold Ca2+ current component recruited by cAMP is selectively associated to the alpha1H (CaV3.2) channel isoform.


Subject(s)
Calcium Channels, T-Type/physiology , Calcium/metabolism , Chromaffin Cells/physiology , Cyclic AMP/pharmacology , Exocytosis/physiology , Ion Channel Gating/physiology , Animals , Calcium Channels, T-Type/drug effects , Cells, Cultured , Chromaffin Cells/drug effects , Differential Threshold/drug effects , Differential Threshold/physiology , Exocytosis/drug effects , Ion Channel Gating/drug effects , Rats , Rats, Sprague-Dawley
12.
J Physiol ; 558(Pt 2): 433-49, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15133061

ABSTRACT

T-type channels are expressed weakly or not at all in adult rat chromaffin cells (RCCs) and there is contrasting evidence as to whether they play a functional role in catecholamine secretion. Here we show that 3-5 days after application of pCPT-cAMP, most RCCs grown in serum-free medium expressed a high density of low-voltage-activated T-type channels without altering the expression and characteristics of high-voltage-activated channels. The density of cAMP-recruited T-type channels increased with time and displayed the typical biophysical and pharmacological properties of low-voltage-activated Ca(2+) channels: (1) steep voltage-dependent activation from -50 mV in 10 mm Ca(2+), (2) slow deactivation but fast and complete inactivation, (3) full inactivation following short conditioning prepulses to -30 mV, (4) effective block of Ca(2+) influx with 50 microM Ni(2+), (5) comparable permeability to Ca(2+) and Ba(2+), and (6) insensitivity to common Ca(2+) channel antagonists. The action of exogenous pCPT-cAMP (200 microM) was prevented by the protein synthesis inhibitor anisomycin and mimicked in most cells by exposure to forskolin and 1-methyl-3-isobutylxanthine (IBMX) or isoprenaline. The protein kinase A (PKA) inhibitor H89 (0.3 microM) and the competitive antagonist of cAMP binding to PKA, Rp-cAMPS, had weak or no effect on the action of pCPT-cAMP. In line with this, the selective Epac agonist 8CPT-2Me-cAMP nicely mimicked the action of pCPT-cAMP and isoprenaline, suggesting the existence of a dominant Epac-dependent recruitment of T-type channels in RCCs that may originate from the activation of beta-adrenoceptors. Stimulation of beta-adrenoceptors occurs autocrinally in RCCs and thus, the neosynthesis of low-voltage-activated channels may represent a new form of 'chromaffin cell plasticity', which contributes, by lowering the threshold of action potential firing, to increasing cell excitability and secretory activity during sustained sympathetic stimulation and/or increased catecholamine circulation.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Calcium Channels, T-Type/physiology , Chromaffin Cells/physiology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Isoproterenol/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Barium/pharmacokinetics , Calcium/pharmacokinetics , Calcium Channel Blockers/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sympathetic Nervous System/physiology
13.
Biophys J ; 85(2): 1326-37, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12885675

ABSTRACT

We have investigated the potentiating action of cAMP on L-currents of rat chromaffin cells and the corresponding increase of Ca(2+)-evoked secretory responses with the aim of separating the action of cAMP on Ca(2+) entry through L-channels and the downstream effects of cAMP/protein kinase A (PKA) on exocytosis. In omega-toxin-treated rat chromaffin cells, exposure to the permeable cAMP analog 8-(4-chlorophenylthio)-adenosine 3',5'-monophosphate (pCPT-cAMP; 1 mM, 30 min) caused a moderate increase of Ca(2+) charge carried through L-channels (19% in 10 mM Ca(2+) at +10 mV) and a drastic potentiation of secretion ( approximately 100%), measured as membrane capacitance increments (deltaC). The apparent Ca(2+) dependency of exocytosis increased with pCPT-cAMP and was accompanied by 83% enhancement of the readily releasable pool of vesicles with no significant change of the probability of release, as evaluated with paired-pulse stimulation protocols. pCPT-cAMP effects could be mimicked by stimulation of beta(1)-adrenoreceptors and reversed by the PKA inhibitor H89, suggesting strict PKA dependence. For short pulses to +10 mV (100 ms), potentiation of exocytosis by pCPT-cAMP was proportional to the quantity of charge entering the cell and occurred independently of whether L, N, or P/Q channels were blocked, suggesting that cAMP acts as a constant amplification factor for secretion regardless of the channel type carrying Ca(2+). Analysis of statistical variations among depolarization-induced capacitance increments indicates that pCPT-cAMP acts downstream of Ca(2+) entry by almost doubling the mean size of unitary exocytic events, most likely as a consequence of an increased granule-to-granule rather than a granule-to-membrane fusion.


Subject(s)
Adaptation, Physiological/physiology , Calcium Channels, L-Type/physiology , Calcium/metabolism , Chromaffin Cells/physiology , Cyclic AMP/metabolism , Exocytosis/physiology , Ion Channel Gating/physiology , Membrane Potentials/physiology , Animals , Cells, Cultured , Female , Rats , Rats, Sprague-Dawley
14.
J Neurosci ; 23(1): 73-83, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12514203

ABSTRACT

Voltage-gated Ca(2+) channels of chromaffin cells are modulated by locally released neurotransmitters through autoreceptor-activated G-proteins. Clear evidence exists in favor of a Ca(2+) channel gating inhibition mediated by purinergic, opioidergic, and alpha-adrenergic autoreceptors. Few and contradictory data suggest also a role of beta-adrenergic autoreceptors (beta-ARs), the action of which, however, remains obscure. Here, using patch-perforated recordings, we show that rat chromaffin cells respond to the beta-AR agonist isoprenaline (ISO) by either upmodulating or downmodulating the amplitude of Ca(2+) currents through two distinct modulatory pathways. ISO (1 microm) could cause either fast inhibition (approximately 25%) or slow potentiation (approximately 25%), or a combination of the two actions. Both effects were completely prevented by propranolol. Slow potentiation was more evident in cells pretreated with pertussis toxin (PTX) or when beta(1)-ARs were selectively stimulated with ISO + ICI118,551. Potentiation was absent when the beta(2)-AR-selective agonist zinterol (1 microm), the protein kinase A (PKA) inhibitor H89, or nifedipine was applied, suggesting that potentiation is associated with a PKA-mediated phosphorylation of L-channels (approximately 40% L-current increase) through beta(1)-ARs. The ISO-induced inhibition was fast and reversible, preserved in cell treated with H89, and mimicked by zinterol. The action of zinterol was mostly on L-channels (38% inhibition). Zinterol action preserved the channel activation kinetics, the voltage-dependence of the I-V characteristic, and was removed by PTX, suggesting that beta(2)AR-mediated channel inhibition was mainly voltage independent and coupled to G(i)/G(o)-proteins. Sequential application of zinterol and ISO mimicked the dual action (inhibition/potentiation) of ISO alone. The two kinetically and pharmacologically distinct beta-ARs signaling uncover alternative pathways, which may serve the autocrine control of Ca(2+)-dependent exocytosis and other related functions of rat chromaffin cells.


Subject(s)
Adrenal Glands/cytology , Calcium Channels, L-Type/physiology , Chromaffin Cells/physiology , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/physiology , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Cells, Cultured , Chromaffin Cells/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Electric Conductivity , Enzyme Inhibitors/pharmacology , Ethanolamines/pharmacology , Female , Heterotrimeric GTP-Binding Proteins/physiology , Isoproterenol/pharmacology , Kinetics , Patch-Clamp Techniques , Pertussis Toxin/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction
15.
Eur J Neurosci ; 16(12): 2297-310, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12492424

ABSTRACT

Chronic application of brain-derived neurotrophic factor (BDNF) induces new selective synthesis of non-L-type Ca2+ channels (N, P/Q, R) at the soma of cultured hippocampal neurons. As N- and P/Q-channels support neurotransmitter release in the hippocampus, this suggests that BDNF-treatment may enhance synaptic transmission by increasing the expression of presynaptic Ca2+ channels as well. To address this issue we studied the long-term effects of BDNF on miniature and stimulus-evoked GABAergic transmission in rat embryo hippocampal neurons. We found that BDNF increased the frequency of miniature currents (mIPSCs) by approximately 40%, with little effects on their amplitude. BDNF nearly doubled the size of evoked postsynaptic currents (eIPSCs) with a marked increase of paired-pulse depression, which is indicative of a major increase in presynaptic activity. The potentiation of eIPSCs was more relevant during the first two weeks in culture, when GABAergic transmission is depolarizing. BDNF action was mediated by TrkB-receptors and had no effects on: (i) the amplitude and dose-response of GABA-evoked IPSCs and (ii) the number of GABA(A) receptor clusters and the total functioning synapses, suggesting that the neurotrophin unlikely acted postsynaptically. In line with this, BDNF affected the contribution of voltage-gated Ca2+ channels mediating evoked GABAergic transmission. BDNF drastically increased the fraction of evoked IPSCs supported by N- and P/Q-channels while it decreased the contribution associated with R- and L-types. This selective action resembles the previously observed up-regulatory effects of BDNF on somatic Ca2+ currents in developing hippocampus, suggesting that potentiation of presynaptic N- and P/Q-channel signalling belongs to a manifold mechanism by which BDNF increases the efficiency of stimulus-evoked GABAergic transmission.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Calcium Channels/metabolism , Hippocampus/embryology , Neural Inhibition/physiology , Presynaptic Terminals/metabolism , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/metabolism , Calcium Channels, P-Type/drug effects , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/drug effects , Calcium Channels, Q-Type/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Female , Fetus , GABA Antagonists/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Neural Inhibition/drug effects , Potassium Chloride/pharmacology , Pregnancy , Presynaptic Terminals/drug effects , Rats , Rats, Sprague-Dawley , Receptor, trkB/drug effects , Receptor, trkB/metabolism , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Synaptic Transmission/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology
16.
Pflugers Arch ; 442(6): 801-13, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11680611

ABSTRACT

Voltage-gated Ca2+ channels are crucial to the control of Ca2+ entry in neurosecretory cells. In the chromaffin cells of adrenal medulla, paracrinally or autocrinally released neurotransmitters induce profound changes in Ca2+ channel gating and Ca2+-dependent events controlling catecholamine secretion and cell activity. The generally held view of these processes is that neurotransmitter-induced modulation of the most widely expressed Ca2+ channels in these cells (N-, P/Q- and L-type) follows two distinct pathways: a direct membrane-delimited Gi/o-protein-induced inhibition of N- and P/Q-type and a remote cAMP-mediated facilitation of L-channels. Both actions depend on voltage, although with remarkably different molecular and kinetic aspects. Recent findings, however, challenge this simple scheme and suggest that L-channels do not require strong pre-pulses to be recruited or facilitated. They are available during normal depolarizations and may be tonically inhibited by Gi/o proteins activated by the released neurotransmitters. Like the N- and P/Q-channels, this autocrine modulation is localized to membrane microareas. Unlike N- and P/Q-channels, however, the inhibition of L-channels is largely independent of voltage and develops in parallel with cAMP-mediated potentiation of channel gating. As L-channels play a crucial role in the control of catecholamine release in chromaffin cells, the two opposite modulations mediated by Gi/o proteins and cAMP may represent an effective way to broaden the dynamic range of Ca2+ signals controlling exocytosis. Here, we review the basic features of this novel L-type channel inhibition comparing it to the well-established forms of L-channel potentiation and voltage-dependent facilitation.


Subject(s)
Calcium Channels, L-Type/physiology , Calcium/metabolism , Cyclic AMP/pharmacology , GTP-Binding Proteins/physiology , Ion Channel Gating , Neurosecretory Systems/metabolism , Animals , Calcium Channels, L-Type/drug effects , Cell Membrane/physiology , Chromaffin Cells/physiology , Electrophysiology , Humans
17.
J Physiol ; 532(Pt 1): 73-90, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11283226

ABSTRACT

Using the cell-attached recording configuration, we found that in adult bovine chromaffin cells there exists a direct membrane-delimited inhibition of single Bay K-modified L-channels mediated by opioids and ATP locally released in the recording pipette. This autocrine modulation is mediated by pertussis toxin (PTX)-sensitive G-proteins and causes a 50 % decrease of the open channel probability (Po) and an equivalent percentage increase of null sweeps at +10 mV with no changes to the activation kinetics, single channel conductance and mean open time. The decrease in Po is mainly due to an increase in the occurrence and duration of slow closed times (> 40 ms). Addition of purinergic and opioidergic antagonists (suramin and naloxone) or cell pre-treatment with PTX removes the inhibition while addition of ATP and opioids inside the pipette, but not outside, mimics the effect. Strong pre-pulses (+150 mV, 280 ms) followed by short repolarizations are unable to remove the inhibition at test potential (+10 mV). Increasing the level of cAMP by either direct application of 8-(4-chlorophenylthio)-cAMP (8-CPT-cAMP) or mixtures of forskolin and 1-methyl-3-isobutylxanthine (IBMX) potentiates the activity of L-channels by increasing the mean open time and decreasing the mean closed time and percentage of null sweeps. The cAMP-induced potentiation occurs regardless of whether the G-protein-mediated inhibition is activated by ATP and opioids or inactivated by PTX. Protein kinase inhibitors (H7 and H89) prevent the effects of cAMP without altering the basal autocrine modulation associated with PTX-sensitive G-proteins. Our results provide new evidence for the coexistence of two distinct modulations that may converge on the same neuroendocrine L-channel: a direct G-protein-dependent inhibition and a cAMP-mediated potentiation, which may work in combination to regulate Ca2+ entry during neurosecretion.


Subject(s)
Autocrine Communication/physiology , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Chromaffin Cells/metabolism , Cyclic AMP/metabolism , Sulfonamides , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Calcium Channels, L-Type/chemistry , Cattle , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Enzyme Inhibitors/pharmacology , GABA Antagonists/pharmacology , Isoquinolines/pharmacology , Kinetics , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Patch-Clamp Techniques , Picrotoxin/pharmacology , Protein Kinase Inhibitors , Suramin/pharmacology
18.
Eur J Neurosci ; 11(10): 3574-84, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10564365

ABSTRACT

The inhibition of L-type channels induced by either bath application of ATP, opioids and catecholamines or by endogenously released neurotransmitters was investigated in rat chromaffin cells with whole-cell recordings (5 mM Ba2+). In both cases, the L-type current, isolated pharmacologically using omega-toxin peptides and potentiated by Bay K 8644, was inhibited by approximately 50% with nearly no changes to the activation-inactivation kinetics. Inhibition was voltage independent at a wide range of potentials (-20 to +50 mV) and insensitive to depolarizing prepulses (+100 mV, 50 ms). Onset and offset of the inhibition were fast (time constants: tau(on) approximately 0.9 s, tau(off) approximately 3.6 s), indicating a rapid mechanism of channel modulation. Whether induced exogenously or from the released granules content in conditions of stopped cell superfusion, the neurotransmitter action was reversible and largely prevented by either intracellular GDP-beta-S, cell treatment with pertussis toxin or simultaneous application of P2y,2x delta/mu-opioidergic and alpha/beta-adrenergic antagonists. This suggests the existence of converging modulatory pathways by which autoreceptors-activated G-proteins reduce the activity of L-type channels through fast interactions. The autocrine inhibition of L-type currents, which was absent in superfused isolated cells, was effective on cell clusters, suggesting that L-type channels may be potently inhibited by cell exocytosis under physiological conditions resembling the intact adrenal glands.


Subject(s)
Adenosine Triphosphate/pharmacology , Analgesics, Opioid/pharmacology , Calcium Channels, L-Type/physiology , Chromaffin Cells/physiology , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Epinephrine/pharmacology , Sympathomimetics/pharmacology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Adrenal Medulla/cytology , Animals , Autocrine Communication/drug effects , Autocrine Communication/physiology , Barium/pharmacokinetics , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Cells, Cultured , Chromaffin Cells/cytology , Chromaffin Cells/drug effects , Electric Stimulation , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Female , GTP-Binding Proteins/physiology , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/pharmacology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Nifedipine/pharmacology , Norepinephrine/pharmacology , Patch-Clamp Techniques , Pertussis Toxin , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic/physiology , Receptors, Purinergic/physiology , Thionucleotides/pharmacology , Virulence Factors, Bordetella/pharmacology
19.
Neuron ; 20(6): 1255-68, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9655512

ABSTRACT

In bovine chromaffin cells, the Ca2+ channels involved in exocytosis are effectively inhibited by ATP and opioids that are coreleased with catecholamines during cell activity. This autocrine loop causes a delay in Ca2+ channel activation that is quickly removed by preceding depolarizations. Changes in Ca2+ channel gating by secreted products thus make it possible to correlate Ca2+ channel activity to secretory events. Here, using cell-attached patch recordings, we found a remarkable correlation between delayed Ca2+ channel openings and neurotransmitter secretion induced by either local or whole-cell Ba2+ stimulation. The action is specific for N- and P/Q-type channels and largely prevented by PTX and mixtures of purinergic and opioid receptor antagonists. Overall, our data provide evidence that exocytosis, viewed through the autocrine inhibition of non-L-type channels, is detectable in membrane patches of approximately 1 microm2 distributed over 30%-40% of the total cell surface, while Ca2+ channels and autoreceptors are uniformly distributed over most of the cell membrane.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium Channels, N-Type , Calcium Channels/metabolism , Chromaffin Cells/metabolism , Ion Channel Gating/physiology , Opioid Peptides/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Adenosine Triphosphate/pharmacology , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Antineoplastic Agents/pharmacology , Autocrine Communication/physiology , Barium/pharmacokinetics , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type , Cattle , Chromaffin Cells/chemistry , Chromaffin Cells/cytology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalin, D-Penicillamine (2,5)- , Enkephalins/pharmacology , Exocytosis/physiology , Ion Channel Gating/drug effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neurotransmitter Agents/metabolism , Nifedipine/pharmacology , Patch-Clamp Techniques , Peptides/pharmacology , Receptors, Opioid/agonists , Receptors, Opioid/metabolism , Receptors, Purinergic/metabolism , Spider Venoms/pharmacology , Suramin/pharmacology , Virulence Factors, Bordetella/pharmacology , omega-Agatoxin IVA , omega-Conotoxin GVIA
20.
J Physiol ; 504 ( Pt 1): 1-15, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9350613

ABSTRACT

1. The Mg2+ block of Na+ and Ca2+ currents through high-voltage activated (HVA; L- and N-type) Ca2+ channels was studied in chick dorsal root ganglion neurones. 2. In low extracellular [Ca2+] (< 10(-8) M) and with Na+o and Cs+i as the main charge carriers (120 mM), HVA Na+ currents started to activate at -40 mV, reached inward peak values near 0 mV and reversed at about +40 mV. 3. Addition of 30-500 microM Mg2+ to the bath caused a strong depression of inward Na+ currents that was voltage and dose dependent (KD = 39 microM in 120 mM Na+ at -10 mV). The block was maximal at negative potentials (< -70 mV) and decreased with increasing positive potentials, suggesting that Mg2+ cannot escape to the cell interior. 4. Block of Ca2+ currents by Mg2+ was also voltage dependent, but by three orders of magnitude less potent than with Na+ currents (KD = 24 mM in 2 mM Ca2+ at -30 mV). The high concentration of Mg2+ caused a prominent voltage shift of channel gating kinetics induced by surface charge screening effects. To compensate for this, Mg2+ block of inward Ca2+ currents was estimated from the instantaneous I-V relationships on return from very positive potentials (+100 mV). 5. Inward Na+ and Ca2+ tail currents following depolarization to +90 mV were markedly depressed, suggesting that channels cleared of Mg2+ ions during strong depolarization are quickly re-blocked on return to negative potentials. The kinetics of re-block by Mg2+ was too fast (< 100 microseconds) to be resolved by our recording apparatus. This implies a rate of entry for Mg2+ > 1.45 x 10(8) M-1 S-1 when Na+ is the permeating ion and a rate approximately 3 orders of magnitude smaller for Ca2+. 6. Mg2+ unblock of HVA Na+ currents at +100 mV was independent of the size of outward currents, whether Na+, Cs+ or NMG+ were the main internal cations. 7. Consistent with the idea of a high-affinity binding site for Ca2+ inside the channel, micromolar amounts of Ca2+ caused a strong depression of Na+ currents between -40 and 0 mV, which was effectively relieved with more positive as well as with negative potentials (KD = 0.7 microM in 120 mM Na+ at -20 mV). In this case, the kinetics of re-block could be resolved and gave rates of entry and exit for Ca2+ of 1.4 x 10(8) M-1 S-1 and 2.95 x 10(2) s-1, respectively. 8. The strong voltage dependence and weak current dependence of HVA channel block by divalent cations and the markedly different KD values of Na+ and Ca2+ current block by Mg2+ can be well described by a previously proposed model for Ca2+ channel permeation based on interactions between the permeating ion and the negative charges forming the high-affinity binding site for Ca2+ inside the pore (Lux, Carbone & Zucker, 1990).


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Calcium/metabolism , Magnesium/pharmacology , Neurons, Afferent/metabolism , Sodium Channel Blockers , Sodium Channels/metabolism , Animals , Chick Embryo , Electric Stimulation , Electrophysiology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ion Channel Gating/drug effects , Kinetics , Membrane Potentials/physiology , Neurons, Afferent/drug effects , Patch-Clamp Techniques , Sodium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...