Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Neurobiol Dis ; 179: 106046, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36806818

RESUMEN

From a pathogenic perspective, Huntington's disease (HD) is being considered as a synaptopathy. As such, alterations in brain neurotransmitter release occur. As the activity of the sympathoadrenal axis is centrally controlled, deficits in the exocytotic release of catecholamine release may also occur. In fact, in chromaffin cells (CCs) of the adrenal medulla of the R6/1 model of HD, decrease of secretion and altered kinetics of the exocytotic fusion pore have been reported. Those alterations could be linked to mitochondrial deficits occurring in peripheral CCs, similar to those described in brain mitochondria. Here we have inquired about alterations in mitochondrial structure and function and their impact on exocytosis and calcium channel currents (ICa). We have monitored various parameters linked to those events, in wild type (WT) and the R6/1 mouse model of HD at a pre-disease stage (2 months age, 2 m), and when motor deficits are present (7 months age, 7 m). In isolated CCs from 7 m and in the adrenal medulla of R6/1 mice, we found the following alterations (with respect 7 m WT mice): (i) augmented fragmented mitochondria and oxidative stress with increased oxidized glutathione; (ii) decreased basal and maximal respiration; (iii) diminution of ATP cell levels; (iv) mitochondrial depolarization; (v) drastic decrease of catecholamine release with poorer potentiation by protonophore FCCP; (vi) decreased ICa inhibition by FCCP; and (vii) lesser potentiation by BayK8644 of ICa and smaller prolongation of current deactivation. Of note was the fact several of these alterations were already manifested in CCs from 2 m R6/1 mice at pre-disease stages. Based on those results, a plausible hypothesis can be raised in the sense that altered mitochondrial function seems to be an early primary event in HD pathogenesis. This is in line with an increasing number of mitochondrial, metabolic, and inflammatory alterations being recently reported in various HD peripheral tissues.


Asunto(s)
Células Cromafines , Enfermedad de Huntington , Ratones , Animales , Enfermedad de Huntington/metabolismo , Calcio/metabolismo , Ratones Transgénicos , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/metabolismo , Células Cromafines/metabolismo , Células Cromafines/patología , Catecolaminas , Mitocondrias/metabolismo , Exocitosis/fisiología , Modelos Animales de Enfermedad
2.
Med Res Rev ; 40(6): 2427-2465, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32677086

RESUMEN

Neurodegenerative diseases (NDDs) represent a huge social burden, particularly in Alzheimer's disease (AD) in which all proposed treatments investigated in murine models have failed during clinical trials (CTs). Thus, novel therapeutic strategies remain crucial. Neuroinflammation is a common pathogenic feature of NDDs. As purinergic P2X7 receptors (P2X7Rs) are gatekeepers of inflammation, they could be developed as drug targets for NDDs. Herein, we review this challenging hypothesis and comment on the numerous studies that have investigated P2X7Rs, emphasizing their molecular structure and functions, as well as their role in inflammation. Then, we elaborate on research undertaken in the field of medicinal chemistry to determine potential P2X7R antagonists. Subsequently, we review the state of neuroinflammation and P2X7R expression in the brain, in animal models and patients suffering from AD, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, multiple sclerosis, and retinal degeneration. Next, we summarize the in vivo studies testing the hypothesis that by mitigating neuroinflammation, P2X7R blockers afford neuroprotection, increasing neuroplasticity and neuronal repair in animal models of NDDs. Finally, we reviewed previous and ongoing CTs investigating compounds directed toward targets associated with NDDs; we propose that CTs with P2X7R antagonists should be initiated. Despite the high expectations for putative P2X7Rs antagonists in various central nervous system diseases, the field is moving forward at a relatively slow pace, presumably due to the complexity of P2X7Rs. A better pharmacological approach to combat NDDs would be a dual strategy, combining P2X7R antagonism with drugs targeting a selective pathway in a given NDD.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Preparaciones Farmacéuticas , Animales , Humanos , Ratones , Enfermedades Neurodegenerativas/tratamiento farmacológico , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7
3.
Int J Mol Sci ; 21(6)2020 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-32178443

RESUMEN

Synaptic disruption and altered neurotransmitter release occurs in the brains of patients and in murine models of neurodegenerative diseases (NDDs). During the last few years, evidence has accumulated suggesting that the sympathoadrenal axis is also affected as disease progresses. Here, we review a few studies done in adrenal medullary chromaffin cells (CCs), that are considered as the amplifying arm of the sympathetic nervous system; the sudden fast exocytotic release of their catecholamines-stored in noradrenergic and adrenergic cells-plays a fundamental role in the stress fight-or-flight response. Bulk exocytosis and the fine kinetics of single-vesicle exocytotic events have been studied in mouse models carrying a mutation linked to NDDs. For instance, in R6/1 mouse models of Huntington's disease (HD), mutated huntingtin is overexpressed in CCs; this causes decreased quantal secretion, smaller quantal size and faster kinetics of the exocytotic fusion pore, pore expansion, and closure. This was accompanied by decreased sodium current, decreased acetylcholine-evoked action potentials, and attenuated [Ca2+]c transients with faster Ca2+ clearance. In the SOD1G93A mouse model of amyotrophic lateral sclerosis (ALS), CCs exhibited secretory single-vesicle spikes with a slower release rate but higher exocytosis. Finally, in the APP/PS1 mouse model of Alzheimer's disease (AD), the stabilization, expansion, and closure of the fusion pore was faster, but the secretion was attenuated. Additionally, α-synuclein that is associated with Parkinson's disease (PD) decreases exocytosis and promotes fusion pore dilation in adrenal CCs. Furthermore, Huntington-associated protein 1 (HAP1) interacts with the huntingtin that, when mutated, causes Huntington's disease (HD); HAP1 reduces full fusion exocytosis by affecting vesicle docking and controlling fusion pore stabilization. The alterations described here are consistent with the hypothesis that central alterations undergone in various NDDs are also manifested at the peripheral sympathoadrenal axis to impair the stress fight-or-flight response in patients suffering from those diseases. Such alterations may occur: (i) primarily by the expression of mutated disease proteins in CCs; (ii) secondarily to stress adaptation imposed by disease progression and the limitations of patient autonomy.


Asunto(s)
Células Cromafines/fisiología , Exocitosis/fisiología , Enfermedades Neurodegenerativas/fisiopatología , Animales , Ratones , Vesículas Secretoras/fisiología , Transmisión Sináptica/fisiología
4.
Pflugers Arch ; 470(8): 1255-1270, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29721607

RESUMEN

Gasotransmitter hydrogen sulphide (H2S) has emerged as a regulator of multiple physiological and pathophysiological processes throughout. Here, we have investigated the effects of NaHS (fast donor of H2S) and GYY4137 (GYY, slow donor of H2S) on the exocytotic release of catecholamines from fast-perifused bovine adrenal chromaffin cells (BCCs) challenged with sequential intermittent pulses of a K+-depolarizing solution. Both donors caused a concentration-dependent facilitation of secretion. This was not due to an augmentation of Ca2+ entry through voltage-activated Ca2+ channels (VACCs) because, in fact, NaHS and GYY caused a mild inhibition of whole-cell Ca2+ currents. Rather, the facilitation of exocytosis seemed to be associated to an augmented basal [Ca2+]c and the K+-elicited [Ca2+]c transients; such effects of H2S donors are aborted by cyclopiazonic acid (CPA), that causes endoplasmic reticulum (ER) Ca2+ depletion through sarcoendoplasmic reticulum Ca2+ ATPase inhibition and by protonophore carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP), that impedes the ability of mitochondria to sequester cytosolic Ca2+ during cell depolarization. Inasmuch as CPA and FCCP reversed the facilitation of secretion triggered by K+ in the presence of NaHS and GYY, is seems that such facilitation is tightly coupled to Ca2+ handling by the ER and mitochondria. On the basis of these results, we propose that H2S regulates catecholamine secretory responses triggered by K+ in BCCs by (i) mobilisation of ER Ca2+ and (ii) interference with mitochondrial Ca2+ circulation. In so doing, the clearance of the [Ca2+]c transient will be delayed and the Ca2+-dependent trafficking of secretory vesicles will be enhanced to overfill the secretory machinery with new vesicles to enhance exocytosis.


Asunto(s)
Calcio/metabolismo , Células Cromafines/efectos de los fármacos , Exocitosis/efectos de los fármacos , Sulfuro de Hidrógeno/farmacología , Animales , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Catecolaminas/metabolismo , Bovinos , Células Cultivadas , Células Cromafines/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Morfolinas/farmacología , Compuestos Organotiofosforados/farmacología , Potasio/metabolismo
5.
J Neurochem ; 147(4): 454-476, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30182387

RESUMEN

As the peripheral sympathoadrenal axis is tightly controlled by the cortex via hypothalamus and brain stem, the central pathological features of Hunting's disease, (HD) that is, deposition of mutated huntingtin and synaptic dysfunctions, could also be expressed in adrenal chromaffin cells. To test this hypothesis we here present a thorough investigation on the pathological and functional changes undergone by chromaffin cells (CCs) from 2-month (2 m) to 7-month (7 m) aged wild-type (WT) and R6/1 mouse model of Huntington's disease (HD), stimulated with acetylcholine (ACh) or high [K+ ] (K+ ). In order to do this, we used different techniques such as inmunohistochemistry, patch-clamp, and amperometric recording. With respect to WT cells, some of the changes next summarized were already observed in HD mice at a pre-disease stage (2 m); however, they were more pronounced at 7 m when motor deficits were clearly established, as follows: (i) huntingtin over-expression as nuclear aggregates in CCs; (ii) smaller CC size with decreased dopamine ß-hydroxylase expression, indicating lesser number of chromaffin secretory vesicles; (iii) reduced adrenal tissue catecholamine content; (iv) reduced Na+ currents with (v) membrane hyperpolarization and reduced ACh-evoked action potentials; (v) reduced [Ca2+ ]c transients with faster Ca2+ clearance; (vi) diminished quantal secretion with smaller vesicle quantal size; (vii) faster kinetics of the exocytotic fusion pore, pore expansion, and closure. On the basis of these data, the hypothesis is here raised in the sense that nuclear deposition of mutated huntingtin in adrenal CCs of R6/1 mice could be primarily responsible for poorer Na+ channel expression and function, giving rise to profound depression of cell excitability, altered Ca2+ handling and exocytosis. OPEN PRACTICES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Cover Image for this issue: doi: 10.1111/jnc.14201.


Asunto(s)
Células Cromafines/metabolismo , Células Cromafines/patología , Exocitosis , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Médula Suprarrenal/metabolismo , Médula Suprarrenal/patología , Animales , Catecolaminas/metabolismo , Humanos , Enfermedad de Huntington/psicología , Cinética , Masculino , Potenciales de la Membrana , Ratones , Ratones Transgénicos , Trastornos del Movimiento/etiología , Trastornos del Movimiento/fisiopatología , Mutación/genética , Desempeño Psicomotor , Canales de Sodio/biosíntesis , Vesículas Sinápticas/patología
6.
Am J Physiol Cell Physiol ; 308(1): C1-19, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25377090

RESUMEN

Altered synaptic transmission with excess glutamate release has been implicated in the loss of motoneurons occurring in amyotrophic lateral sclerosis (ALS). Hyperexcitability or hypoexcitability of motoneurons from mice carrying the ALS mutation SOD1(G93A) (mSOD1) has also been reported. Here we have investigated the excitability, the ion currents, and the kinetics of the exocytotic fusion pore in chromaffin cells from postnatal day 90 to postnatal day 130 mSOD1 mice, when motor deficits are already established. With respect to wild-type (WT), mSOD1 chromaffin cells had a decrease in the following parameters: 95% in spontaneous action potentials, 70% in nicotinic current for acetylcholine (ACh), 35% in Na(+) current, 40% in Ca(2+)-dependent K(+) current, and 53% in voltage-dependent K(+) current. Ca(2+) current was increased by 37%, but the ACh-evoked elevation of cytosolic Ca(2+) was unchanged. Single exocytotic spike events triggered by ACh had the following differences (mSOD1 vs. WT): 36% lower rise rate, 60% higher decay time, 51% higher half-width, 13% lower amplitude, and 61% higher quantal size. The expression of the α3-subtype of nicotinic receptors and proteins of the exocytotic machinery was unchanged in the brain and adrenal medulla of mSOD1, with respect to WT mice. A slower fusion pore opening, expansion, and closure are likely linked to the pronounced reduction in cell excitability and in the ion currents driving action potentials in mSOD1, compared with WT chromaffin cells.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Catecolaminas/metabolismo , Células Cromafines/enzimología , Exocitosis , Fusión de Membrana , Superóxido Dismutasa/metabolismo , Transmisión Sináptica , Acetilcolina/farmacología , Potenciales de Acción , Factores de Edad , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Calcio/metabolismo , Señalización del Calcio , Células Cromafines/efectos de los fármacos , Células Cromafines/metabolismo , Modelos Animales de Enfermedad , Exocitosis/efectos de los fármacos , Humanos , Transporte Iónico , Cinética , Masculino , Fusión de Membrana/efectos de los fármacos , Ratones Transgénicos , Actividad Motora , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Mutación , Potasio/metabolismo , Receptores Nicotínicos/efectos de los fármacos , Receptores Nicotínicos/metabolismo , Sodio/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Transmisión Sináptica/efectos de los fármacos
7.
J Neurochem ; 125(2): 205-13, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23350646

RESUMEN

Chondroitin sulfate (CS) proteoglycans (CSPGs) are the most abundant PGs of the brain extracellular matrix (ECM). Free CS could be released during ECM degradation and exert physiological functions; thus, we aimed to investigate the effects of CS on voltage- and current-clamped rat embryo hippocampal neurons in primary cultures. We found that CS elicited a whole-cell Na(+)-dependent inward current (ICS) that produced drastic cell depolarization, and a cytosolic calcium transient ([Ca(2+)]c). Those effects were similar to those elicited by α-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA) and kainate, were completely blocked by NBQX and CNQX, were partially blocked by GYKI, and were unaffected by MK801 and D-APV. Furthermore, ICS and AMPA currents were similarly potentiated by cyclothiazide, a positive allosteric modulator of AMPA receptors. Because CSPGs have been attributed Ca(2) (+) -dependent roles, such as neural network development, axon pathfinding, plasticity and regeneration after CNS injury, CS action after ECM degradation could be contributing to the mediation of these effects through its interaction with AMPA and kainate receptors.


Asunto(s)
Potenciales de Acción/fisiología , Sulfatos de Condroitina/metabolismo , Neuronas/metabolismo , Receptores AMPA/metabolismo , Receptores de Ácido Kaínico/metabolismo , Animales , Células Cultivadas , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Hipocampo/metabolismo , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
8.
Pflugers Arch ; 464(1): 33-41, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22278417

RESUMEN

Chromaffin cells are an excellent model for stimulus-secretion coupling. Ca(2+) entry through plasma membrane voltage-operated Ca(2+) channels (VOCC) is the trigger for secretion, but the intracellular organelles contribute subtle nuances to the Ca(2+) signal. The endoplasmic reticulum amplifies the cytosolic Ca(2+) ([Ca(2+)](C)) signal by Ca(2+)-induced Ca(2+) release (CICR) and helps generation of microdomains with high [Ca(2+)](C) (HCMD) at the subplasmalemmal region. These HCMD induce exocytosis of the docked secretory vesicles. Mitochondria close to VOCC take up large amounts of Ca(2+) from HCMD and stop progression of the Ca(2+) wave towards the cell core. On the other hand, the increase of [Ca(2+)] at the mitochondrial matrix stimulates respiration and tunes energy production to the increased needs of the exocytic activity. At the end of stimulation, [Ca(2+)](C) decreases rapidly and mitochondria release the Ca(2+) accumulated in the matrix through the Na(+)/Ca(2+) exchanger. VOCC, CICR sites and nearby mitochondria form functional triads that co-localize at the subplasmalemmal area, where secretory vesicles wait ready for exocytosis. These triads optimize stimulus-secretion coupling while avoiding propagation of the Ca(2+) signal to the cell core. Perturbation of their functioning in neurons may contribute to the genesis of excitotoxicity, ageing mental retardation and/or neurodegenerative disorders.


Asunto(s)
Calcio/metabolismo , Células Cromafines/metabolismo , Exocitosis , Mitocondrias/metabolismo , Animales , Canales de Calcio/metabolismo , Citosol/metabolismo , Humanos , Vesículas Secretoras/metabolismo
9.
Biochem Biophys Res Commun ; 428(4): 482-6, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23123627

RESUMEN

The kinetics of single-amperometric exocytotic events has been measured in chromaffin cells of C57 mice and in an APP/PS1 mouse model of Alzheimer's disease (AD). K(+) depolarisation causes a burst of spikes that indicate the quantal release of the single-vesicle content of catecholamine. The kinetic analysis of 278 spikes from 10 control cells and 520 spikes from 18 APP/PS1 cells shows the following features of the latter compared with the former: (i) 45% lower t(1/2); (ii) 60% smaller quantal size; (iii) 50% lower decay time. Spike feet also showed 60% smaller quantal size. Immunofluorescence and thioflavin staining showed no amyloid beta (Aß) burden in adrenal medulla slices of APP/PS1 mice that however exhibited dense Aß plaques in the cortex and hippocampus. Furthermore, acetylcholinesterase staining of adrenal medulla indicated no apparent differences in the innervation by splanchnic cholinergic nerve terminals of chromaffin cells from control and APP/PS1 mice. This is the first report identifying subtle differences in the last steps of exocytosis that could be an indication of synaptic dysfunction of the secretory machinery not linked to Aß burden in AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Catecolaminas/metabolismo , Células Cromafines/metabolismo , Exocitosis , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Presenilina-1/genética , Nervios Esplácnicos/fisiopatología
10.
J Med Chem ; 64(4): 2272-2290, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33560845

RESUMEN

The ATP-gated P2X7 purinergic receptor (P2X7) is involved in the pathogenesis of many neurodegenerative diseases (NDDs). Several P2X7 antagonists have been developed, though none of them reached clinical trials for this indication. In this work, we designed and synthesized novel blood-brain barrier (BBB)-permeable derivatives as potential P2X7 antagonists. They comprise purine or xanthine cores linked to an aryl group through different short spacers. Compounds were tested in YO-PRO-1 uptake assays and intracellular calcium dynamics in a human P2X7-expressing HEK293 cell line, two-electrode voltage-clamp recordings in Xenopus laevis oocytes, and in interleukin 1ß release assays in mouse peritoneal macrophages. BBB permeability was assessed by parallel artificial membrane permeability assays and P-glycoprotein ATPase activity. Dichloroarylpurinylethanones featured a certain P2X7 blockade, being compound 6 (2-(6-chloro-9H-purin-9-yl)-1-(2,4-dichlorophenyl)ethan-1-one), named ITH15004, the most potent, selective, and BBB-permeable antagonist. Compound 6 can be considered as a first non-nucleotide purine hit for future drug optimizations.


Asunto(s)
Antiinflamatorios/farmacología , Antagonistas del Receptor Purinérgico P2X/farmacología , Purinas/farmacología , Receptores Purinérgicos P2X7/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Antiinflamatorios/síntesis química , Antiinflamatorios/metabolismo , Células HEK293 , Humanos , Interleucina-1beta/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Oocitos/efectos de los fármacos , Antagonistas del Receptor Purinérgico P2X/síntesis química , Antagonistas del Receptor Purinérgico P2X/metabolismo , Purinas/síntesis química , Purinas/metabolismo , Xenopus laevis
11.
Am J Physiol Cell Physiol ; 298(2): C397-405, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19940070

RESUMEN

The ability of adrenal chromaffin cells to fast-release catecholamines relies on their capacity to fire action potentials (APs). However, little attention has been paid to the requirements needed to evoke the controlled firing of APs. Few data are available in rodents and none on the bovine chromaffin cell, a model extensively used by researchers. The aim of this work was to clarify this issue. Short puffs of acetylcholine (ACh) were fast perifused to current-clamped chromaffin cells and produced the firing of single APs. Based on the currents generated by such ACh applications and previous literature, current waveforms that efficiently elicited APs at frequencies up to 20 Hz were generated. Complex waveforms were also generated by adding simple waveforms with different delays; these waveforms aimed at modeling the stimulation patterns that a chromaffin cell would conceivably undergo upon strong synaptic stimulation. Cholinergic innervation was assessed using the acetylcholinesterase staining technique on the supposition that the innervation pattern is a determinant of the kind of stimuli chromaffin cells can receive. It is concluded that 1) a reliable method to produce frequency-controlled APs by applying defined current injection waveforms is achieved; 2) the APs thus generated have essentially the same features as those spontaneously emitted by the cell and those elicited by fast-ACh perifusion; 3) the higher frequencies attainable peak at around 30 Hz; and 4) the bovine adrenal medulla shows abundant cholinergic innervation, and chromaffin cells show strong acetylcholinesterase staining, consistent with a tight cholinergic presynaptic control of firing frequency.


Asunto(s)
Acetilcolina/metabolismo , Médula Suprarrenal/inervación , Catecolaminas/metabolismo , Fibras Colinérgicas/metabolismo , Células Cromafines/metabolismo , Nervios Esplácnicos/metabolismo , Transmisión Sináptica , Acetilcolinesterasa/metabolismo , Animales , Bovinos , Células Cultivadas , Células Cromafines/enzimología , Estimulación Eléctrica , Potenciales Evocados , Femenino , Cinética , Técnicas de Placa-Clamp , Terminales Presinápticos/metabolismo
12.
Mol Pharmacol ; 78(4): 734-44, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20631052

RESUMEN

The cardiovascular protecting effects of resveratrol, an antioxidant polyphenol present in grapes and wine, have been attributed to its vasorelaxing effects and to its anti-inflammatory, antioxidant, and antiplatelet actions. Inhibition of adrenal catecholamine release has also been recently implicated in its cardioprotecting effects. Here, we have studied the effects of nanomolar concentrations of resveratrol on quantal single-vesicle catecholamine release in isolated bovine adrenal chromaffin cells. We have found that 30 to 300 nM concentrations of resveratrol blocked the acetylcholine (ACh) and high K(+)-evoked quantal catecholamine release, amperometrically measured with a carbon fiber microelectrode. At these concentrations, resveratrol did not affect the whole-cell inward currents through nicotinic receptors or voltage-dependent sodium and calcium channels, neither the ACh- or K(+)-elicited transients of cytosolic Ca(2+). Blockade by nanomolar resveratrol of secretion in ionomycin- or digitonin-treated cells suggests an intracellular site of action beyond Ca(2+)-dependent exocytotic steps. The fact that nanomolar resveratrol augmented cGMP is consistent with the view that resveratrol could be blocking the quantal secretion of catecholamine through a nitric oxide-linked mechanism. Because this effect occurs at nanomolar concentrations, our data are relevant in the context of the low circulating levels of resveratrol found in moderate consumers of red wines, which could afford cardioprotection by mitigating the catecholamine surge occurring during stress.


Asunto(s)
Médula Suprarrenal/citología , Catecolaminas/antagonistas & inhibidores , Catecolaminas/metabolismo , Células Cromafines/efectos de los fármacos , Células Cromafines/metabolismo , Estilbenos/administración & dosificación , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Médula Suprarrenal/efectos de los fármacos , Médula Suprarrenal/metabolismo , Animales , Bovinos , Células Cultivadas , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Procedimientos Analíticos en Microchip/métodos , Resveratrol
13.
Nat Commun ; 11(1): 3208, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32587250

RESUMEN

Inner hair cells (IHCs) are the primary receptors for hearing. They are housed in the cochlea and convey sound information to the brain via synapses with the auditory nerve. IHCs have been thought to be electrically and metabolically independent from each other. We report that, upon developmental maturation, in mice 30% of the IHCs are electrochemically coupled in 'mini-syncytia'. This coupling permits transfer of fluorescently-labeled metabolites and macromolecular tracers. The membrane capacitance, Ca2+-current, and resting current increase with the number of dye-coupled IHCs. Dual voltage-clamp experiments substantiate low resistance electrical coupling. Pharmacology and tracer permeability rule out coupling by gap junctions and purinoceptors. 3D electron microscopy indicates instead that IHCs are coupled by membrane fusion sites. Consequently, depolarization of one IHC triggers presynaptic Ca2+-influx at active zones in the entire mini-syncytium. Based on our findings and modeling, we propose that IHC-mini-syncytia enhance sensitivity and reliability of cochlear sound encoding.


Asunto(s)
Cóclea , Células Ciliadas Auditivas Internas , Audición/fisiología , Animales , Señalización del Calcio , Cóclea/citología , Cóclea/inervación , Nervio Coclear/metabolismo , Tomografía con Microscopio Electrónico , Células Gigantes , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Internas/fisiología , Ratones , Técnicas de Placa-Clamp , Roedores/fisiología , Sinapsis/metabolismo
14.
Am J Physiol Cell Physiol ; 297(2): C407-18, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19439528

RESUMEN

Studies on the bulk catecholamine release from fetal and neonatal rat adrenals, adrenal slices, or isolated chromaffin cells stimulated with high K(+), hypoxia, hypercapnia, or acidosis are available. However, a study analyzing the kinetics of quantal secretion is lacking. We report here such a study in which we compare the quantal release of catecholamines from immature rat embryo chromaffin cells (ECCs) and their mothers' (MCCs). Cell challenging with a strong depolarizing stimulus (75 mM K(+)) caused spike bursts having the following characteristics. ECCs released more multispike events and wave envelopes than MCCs. This, together with narrower single-spike events, a faster decay, and a threefold smaller quantal size suggest a faster secretory machinery in ECCs. Furthermore, with a milder stimulus (25 mM K(+)) enhanced Ca(2+) entry by L-type Ca(2+) channel activator BAY K 8644 did not change the kinetic parameters of single spikes in ECCs; in contrast, augmentation of Ca(2+) entry increased spike amplitude and width, quantal size, and decay time in MCCs. This suggests that in mature MCCs, the last exocytotic steps are more tightly regulated than in immature ECCs. Finally, we found that quantal secretion was fully controlled by L-type voltage-dependent Ca(2+) channels (VDCCs) in ECCs, whereas both L- and non-L VDCCs (N and PQ) contributed equally to secretion control in MCCs. Our results have the following physiological, pharmacological, and clinical relevance: 1) they may help to better understand the regulation of adrenal catecholamine release in response to stress during fetal life and delivery; 2) if clinically used, L-type Ca(2+) channel blockers may augment the incidence of sudden infant death syndrome (SIDS); and 3) so-called Ca(2+) promotors or activators of Ca(2+) entry through L-type VDCCs may be useful to secure a healthy catecholamine surge upon violent stress during fetal life, at birth, or to prevent the SIDS in neonates at risk.


Asunto(s)
Catecolaminas/metabolismo , Células Cromafines/metabolismo , Embrión de Mamíferos , Ácido 3-piridinacarboxílico, 1,4-dihidro-2,6-dimetil-5-nitro-4-(2-(trifluorometil)fenil)-, Éster Metílico/metabolismo , Médula Suprarrenal/citología , Animales , Agonistas de los Canales de Calcio/metabolismo , Bloqueadores de los Canales de Calcio/metabolismo , Canales de Calcio/metabolismo , Células Cromafines/citología , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Humanos , Nimodipina/metabolismo , Potasio/metabolismo , Embarazo , Ratas , Ratas Wistar , omega-Conotoxinas/metabolismo
15.
J Pharmacol Exp Ther ; 330(3): 844-54, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19509314

RESUMEN

Mitochondrial calcium (Ca(2+)) dyshomeostasis constitutes a critical step in the metabolic crossroads leading to cell death. Therefore, we have studied here whether 7-chloro-5-(2-chlorophenyl)-1,5-dihydro-4,1-benzothiazepin-2(3H)-one (CGP37157; CGP), a blocker of the mitochondrial Na(+)/Ca(2+)-exchanger (mNCX), protects against veratridine-elicited chromaffin cell death, a model suitable to study cell death associated with Ca(2+) overload. Veratridine produced a concentration-dependent cell death, measured as lactate dehydrogenase released into the medium after a 24-h incubation period. CGP rescued cells from veratridine-elicited death in a concentration-dependent manner; its EC(50) was approximately 10 microM, and 20 to 30 microM caused near 100% cytoprotection. If preincubated for 30 min and washed out for 3 min before adding veratridine, CGP still afforded significant cytoprotection. At 30 microM, CGP blocked the veratridine-elicited free radical production, mitochondrial depolarization, and cytochrome c release. At this concentration, CGP also inhibited the Na(+) and Ca(2+) currents by 50 to 60% and the veratridine-elicited oscillations of cytosolic Ca(2+). This drastic cytoprotective effect of CGP could be explained in part through its regulatory actions on the mNCX.


Asunto(s)
Células Cromafines/efectos de los fármacos , Clonazepam/análogos & derivados , Mitocondrias/metabolismo , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Tiazepinas/farmacología , Veratridina/antagonistas & inhibidores , Veratridina/toxicidad , Animales , Canales de Calcio/efectos de los fármacos , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Bovinos , Muerte Celular/efectos de los fármacos , Células Cultivadas , Células Cromafines/enzimología , Clonazepam/farmacología , Colorantes , Citocromos c/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Técnicas de Placa-Clamp , Especies Reactivas de Oxígeno , Canales de Sodio/efectos de los fármacos , Canales de Sodio/metabolismo , Sales de Tetrazolio , Tiazoles
16.
Neurotoxicology ; 70: 99-111, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30448301

RESUMEN

In the frame of a repositioning programme with cholinergic medicines in clinical use searching for neuroprotective properties, we surprisingly found that spasmolytic antimuscarinics otilonium and pinaverium exhibited neurotoxic effects in neuronal cultures. We decided to characterize such unexpected action in primary cultures of rat embryo cortical neurons. Neurotoxicity was time- and concentration-dependent, exhibiting approximate EC50 values of 5 µM for both drugs. Seven antimuscarinic drugs endowed with a quaternary ammonium, and another 10 drugs with different cholinergic activities, carrying in their molecule a ternary ammonium did not exhibit neurotoxicity. Both drugs caused a concentration-dependent blockade of whole-cell inward currents through voltage-activated calcium channels (VACCs). Consistent with this, they also blocked the K+-elicited [Ca2+]c transients. Neither antioxidant catalase, glutathione, n-acetylcysteine, nor melatonin protected against neurotoxicity of otilonium or pinaverium. However cyclosporine A, a blocker of the mitochondrial permeability transition pore, prevented the neurotoxic effects of otilonium and pinaverium monitored as the fraction of cells undergoing apoptosis. Furthermore, the caspase-9 and caspase-3 inhibitor Ac-LEHD-CHO mitigated the apoptotic neuronal death of both drugs by around 50%. Data are compatible with the hypothesis that otilonium and pinaverium elicit neuronal death by activating the intrinsic mitochondrial-mediated signaling pathway of apoptosis. This may have its origin in the mitigation of Ca2+ entry and the uncoupling of the Ca2+-dependent generation of mitochondrial bioenergetics, thus causing the opening of the mitochondrial mPTP to elicit apoptotic neuronal death.


Asunto(s)
Apoptosis/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Morfolinas/toxicidad , Neuronas/efectos de los fármacos , Compuestos de Amonio Cuaternario/toxicidad , Animales , Apoptosis/fisiología , Bovinos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Corteza Cerebral/patología , Corteza Cerebral/fisiología , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Femenino , Humanos , Mitocondrias/patología , Mitocondrias/fisiología , Antagonistas Muscarínicos , Neuronas/patología , Neuronas/fisiología , Embarazo , Ratas , Ratas Sprague-Dawley
17.
Biochem Biophys Res Commun ; 365(3): 413-9, 2008 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-17981151

RESUMEN

The view that Ca(2+) entry through voltage-dependent Ca(2+) channels (VDCC) and through nicotinic receptors for acetylcholine (nAChRs) causes equal catecholamine release responses in chromaffin cells, was reinvestigated here using new protocols. We have made two-step experiments consisting in an ACh prepulse followed by a depolarizing pulse (DP). In voltage-clamped bovine chromaffin cells an ACh prepulse caused a slow-rate release but augmented 4.5-fold the much faster exocytotic response triggered by a subsequent depolarizing pulse (measured with capacitance and amperometry). If the ACh prepulse was given with mecamylamine or in low external Ca(2+), the secretion increase disappeared. This suggests a two-step model for the effects of ACh: (1) meager Ca(2+) entry through nAChRs mostly serves to keep loaded with vesicles the secretory machine; and (2) in this manner, the cell is prepared to respond with an explosive secretion of catecholamine upon depolarization and fast high Ca(2+) entry through VDCC.


Asunto(s)
Acetilcolina/metabolismo , Calcio/metabolismo , Catecolaminas/metabolismo , Células Cromafines/metabolismo , Exocitosis , Receptores Nicotínicos/metabolismo , Animales , Canales de Calcio/metabolismo , Bovinos , Potenciales Evocados , Modelos Biológicos , Técnicas de Placa-Clamp
18.
J Pharmacol Exp Ther ; 324(2): 685-93, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17962518

RESUMEN

In a previous study performed in the intact adrenal gland (Lim et al., 2002), stimulation with acetylcholine (ACh) or high K(+) concentrations (K(+)) produced greater catecholamine release in spontaneously hypertensive rats (SHR), as compared with normotensive animals. In this study, the time course of secretion was in the range of minutes. Hence, we do not know whether enhanced release is due to greater quantal content and/or distinct kinetics in SHRs and control animals. To get insight into the mechanism involved in such enhanced catecholamine secretory responses, we performed a single-vesicle release study in primary cultures of adrenal chromaffin cells, recorded with amperometry. Cells were stimulated with 2-s pulses of 1 mM ACh or 70 mM K(+). The secretory responses to ACh or K(+) pulses in SHR cells as compared with control cells had the following characteristics: 1) double number of secretory events, 2) 4-fold augmentation of total secretion, 3) cumulative secretion that saturated slowly, 4) 3-fold higher complex events with two to four superimposed spikes that may be explained by faster spike kinetics, 5) about 2- to 3-fold higher event frequency at earlier post stimulation periods, and 6) 2- to 5-fold higher quantal content of simple spikes. We conclude that SHR cells have faster and larger catecholamine release responses, explained by more vesicles ready to undergo exocytosis and greater quantal content of vesicles. This could have relevance to further understand the pathogenic mechanisms involved in the development of high blood pressure, as well as in the identification of new drug targets to treat hypertension.


Asunto(s)
Catecolaminas/metabolismo , Catecolaminas/farmacocinética , Células Cromafines/metabolismo , Hipertensión/metabolismo , Vesículas Secretoras/metabolismo , Potenciales de Acción/fisiología , Animales , Presión Sanguínea/fisiología , Exocitosis/fisiología , Masculino , Ratas , Ratas Endogámicas SHR , Ratas Sprague-Dawley
19.
Eur J Pharmacol ; 598(1-3): 81-6, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-18831972

RESUMEN

Tobacco smokers have an increased risk of cardiovascular disease; this is likely associated to an enhanced catecholamine release by circulating nicotine. Here, we have explored how low concentrations of nicotine in the range of those found in the blood of tobacco smokers, might affect the release of catecholamines in bovine chromaffin cells. We have combined patch-clamp and Ca(2+) imaging techniques to study cell excitability, cytosolic Ca(2+) transients, vesicle movement, and secretory responses. We found that low concentrations of nicotine (1.5-3 microM) did not enhance catecholamine release by themselves. However, they drastically augmented the catecholamine release response triggered by a supramaximal K(+) depolarising pulse. Furthermore, low nicotine concentrations caused slight depolarisation with superimposed action potentials, a transient elevation of [Ca(2+)](c) and augmented Ca(2+)-dependent vesicle motion underneath the plasmalemma. We suggest that low nicotine concentrations overload the secretory machinery with secretory vesicles, which cause chromaffin cells to respond with an exaggerated adrenaline release into the circulation during stress. This might contribute to the higher cardiovascular risk of tobacco smokers.


Asunto(s)
Células Cromafines/efectos de los fármacos , Vesículas Citoplasmáticas/efectos de los fármacos , Exocitosis/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Potasio/farmacología , Potenciales de Acción/efectos de los fármacos , Compuestos de Anilina , Animales , Catecolaminas/metabolismo , Bovinos , Separación Celular , Sinergismo Farmacológico , Electrofisiología , Colorantes Fluorescentes , Potenciales de la Membrana/efectos de los fármacos , Microscopía Fluorescente , Xantenos
20.
Curr Mol Pharmacol ; 8(1): 81-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25966701

RESUMEN

Here we review the contribution of the various subtypes of voltage-activated calcium channels (VACCs) to the regulation of catecholamine release from chromaffin cells (CCs) at early life. Patch-clamp recording of inward currents through VACCs has revealed the expression of high-threshold VACCs (high-VACCs) of the L, N, and PQ subtypes in rat embryo CCs and ovine embryo CCs. Low-threshold VACC (low-VACC) currents (T-type) have also been recorded in rat embryo CCs and rat neonatal slices of adrenal medullae. Near full blockade by nifedipine and nimodipine of the K(+)-elicited secretion as well as the hypoxia induced secretion (HIS) supports the dominant role of L-VACC subtypes to the regulation of exocytosis at early life. Partial blockade by ω-conotoxin GVIA and ω-agatoxin IVA suggests a transient participation of N and PQ high-VACCs to the regulation of the HIS response at early stages of CC exposure to hypoxia. T-type low-VACC current did not elicit exocytosis triggered by electrical depolarising pulses applied to rat embryo CCs in one study, but largely contributed to the HIS response in neonatal rat adrenal slices in another. In spite of scarce available data, the sequence of events driving the HIS response in CCs at early life could be established as follows: (i) hypoxia blocks one or more K(+) channels; (ii) as a consequence, mild membrane depolarisation occurs; (iii) T-type low-VACCs open at membrane potentials more hyperpolarised than those required to recruit the high-VACCs; (iv) firing of action potentials then occurs; (v) fast-inactivating N and PQ high-VACCs transiently open and low-inactivating L high-VACCs remain open along the hypoxia stimulus; (vi) increase of cytosolic Ca(2+) takes place; and (vii) the exocytotic release of catecholamine occurs in two phases, an explosive initial phase, driven by Ca(2+) entry through L, N and PQ channels, followed by a more sustained catecholamine release at a slower rate driven by L-type channels.


Asunto(s)
Canales de Calcio/metabolismo , Células Cromafines/metabolismo , Exocitosis , Animales , Canales de Calcio/clasificación , Humanos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA