Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
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.
Biochim Biophys Acta ; 1828(7): 1608-18, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23159773

ABSTRACT

Voltage-gated Ca²âº channels (VGCCs) are voltage sensors that convert membrane depolarizations into Ca²âº signals. In the chromaffin cells of the adrenal medulla, the Ca²âº signals driven by VGCCs regulate catecholamine secretion, vesicle retrievals, action potential shape and firing frequency. Among the VGCC-types expressed in these cells (N-, L-, P/Q-, R- and T-types), the two L-type isoforms, Ca(v)1.2 and Ca(v)1.3, control key activities due to their particular activation-inactivation gating and high-density of expression in rodents and humans. The two isoforms are also effectively modulated by G protein-coupled receptor pathways delimited in membrane micro-domains and by the cAMP/PKA and NO/cGMP/PKG phosphorylation pathways which induce prominent Ca²âº current changes if opposingly regulated. The two L-type isoforms shape the action potential and directly participate to vesicle exocytosis and endocytosis. The low-threshold of activation and slow rate of inactivation of Ca(v)1.3 confer to this channel the unique property of carrying sufficient inward current at subthreshold potentials able to activate BK and SK channels which set the resting potential, the action potential shape, the cell firing mode and the degree of spike frequency adaptation during spontaneous firing or sustained depolarizations. These properties help chromaffin cells to optimally adapt when switching from normal to stress-mimicking conditions. Here, we will review past and recent findings on cAMP- and cGMP-mediated modulations of Ca(v)1.2 and Ca(v)1.3 and the role that these channels play in the control of chromaffin cell firing. This article is part of a Special Issue entitled: Calcium channels.


Subject(s)
Adrenal Glands/metabolism , Biological Clocks/physiology , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Chromaffin Cells/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Action Potentials , Animals , Exocytosis , Humans , Phosphorylation
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...