Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 179: 106046, 2023 04.
Article in English | MEDLINE | ID: mdl-36806818

ABSTRACT

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.


Subject(s)
Chromaffin Cells , Huntington Disease , Mice , Animals , Huntington Disease/metabolism , Calcium/metabolism , Mice, Transgenic , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/metabolism , Chromaffin Cells/metabolism , Chromaffin Cells/pathology , Catecholamines , Mitochondria/metabolism , Exocytosis/physiology , Disease Models, Animal
3.
Pflugers Arch ; 473(2): 273-286, 2021 02.
Article in English | MEDLINE | ID: mdl-33108514

ABSTRACT

Pre-blockade of the sarco-endoplasmic reticulum (ER) calcium ATPase (SERCA) with irreversible thapsigargin depresses exocytosis in adrenal bovine chromaffin cells (BCCs). Distinct expression of voltage-dependent Ca2+-channel subtypes and of the Ca2+-induced Ca2+ release (CICR) mechanism in BCCs versus mouse chromaffin cells (MCCs) has been described. We present a parallel study on the effects of the acute SERCA blockade with reversible cyclopizonic acid (CPA), to repeated pulsing with acetylcholine (ACh) at short (15 s) and long intervals (60 s) at 37 °C, allowing the monitoring of the initial size of a ready-release vesicle pool (RRP) and its depletion and recovery in subsequent stimuli. We found (i) strong depression of exocytosis upon ACh pulsing at 15-s intervals and slower depression at 60-s intervals in both cell types; (ii) facilitation of exocytosis upon acute SERCA inhibition, with back to depression upon CPA washout in MCCs; (iii) blockade of exocytosis upon acute SERCA inhibition and pronounced rebound of exocytosis upon CPA washout in BCCs; (iv) basal [Ca2+]c elevation upon stimulation with ACh at short intervals (but not at long intervals) in both cell types; and (v) augmentation of basal [Ca2+]c and inhibition of peak [Ca2+]c amplitude upon CPA treatment in both cell types, with milder effects upon stimulation at 60-s intervals. These results are compatible with the view that while in MCCs the uptake of Ca2+ via SERCA contributes to the mitigation of physiological ACh triggered secretion, in BCCs the uptake of Ca2+ into the ER facilitates such responses likely potentiating a Ca2+-induced Ca2+ release mechanism. These drastic differences in the regulation of ACh-triggered secretion at 37 °C may help to understand different patterns of the regulation of exocytosis by the circulation of Ca2+ at a functional ER Ca2+ store.


Subject(s)
Calcium/metabolism , Catecholamines/metabolism , Chromaffin Cells/drug effects , Endoplasmic Reticulum/drug effects , Enzyme Inhibitors/pharmacology , Exocytosis/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Acetylcholine/pharmacology , Animals , Calcium Signaling/drug effects , Cattle , Cells, Cultured , Chromaffin Cells/enzymology , Endoplasmic Reticulum/enzymology , Indoles/pharmacology , Male , Mice, Inbred C57BL , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Species Specificity , Thapsigargin/pharmacology , Time Factors
4.
J Neurochem ; 147(4): 454-476, 2018 11.
Article in English | MEDLINE | ID: mdl-30182387

ABSTRACT

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.


Subject(s)
Chromaffin Cells/metabolism , Chromaffin Cells/pathology , Exocytosis , Huntingtin Protein/biosynthesis , Huntingtin Protein/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Adrenal Medulla/metabolism , Adrenal Medulla/pathology , Animals , Catecholamines/metabolism , Humans , Huntington Disease/psychology , Kinetics , Male , Membrane Potentials , Mice , Mice, Transgenic , Movement Disorders/etiology , Movement Disorders/physiopathology , Mutation/genetics , Psychomotor Performance , Sodium Channels/biosynthesis , Synaptic Vesicles/pathology
5.
J Pharmacol Exp Ther ; 367(1): 28-39, 2018 10.
Article in English | MEDLINE | ID: mdl-30006476

ABSTRACT

The inhibition of nicotinic acetylcholine receptors (nAChRs) has been proposed as a potential strategy to develop new antidepressant drugs. This is based on the observation that antidepressants that selectively block noradrenaline (NA) or serotonin (5-HT) reuptake also inhibit nAChRs. Dual antidepressants blocking both NA and 5-HT reuptake were proposed to shorten the delay in exerting their clinical effects; whether duloxetine, a prototype of dual antidepressants, also blocks nAChRs is unknown. Here we explored this question in bovine chromaffin cells (BCCs) that express native α3, α5, and α7 nAChRs and in cell lines expressing human α7, α3ß4, or α4ß2 nAChRs. We have found that duloxetine fully blocked the acetylcholine (ACh)-elicited nicotinic currents in BCCs with an IC50 of 0.86 µM. Such blockade seemed to be noncompetitive, voltage dependent, and partially use dependent. The ACh-elicited membrane depolarization, the elevation of cytosolic calcium ([Ca2+]c), and catecholamine release in BCCs were also blocked by duloxetine. This blockade developed slowly, and the recovery of secretion was also slow and gradual. Duloxetine did not affect Na+ or Ca2+ channel currents neither the high-K+-elicited [Ca2+]c transients and secretion. Of interest was that in cell lines expressing human α7, α3ß4, and α4ß2 nAChRs, duloxetine blocked nicotinic currents with IC50 values of 0.1, 0.56, and 0.85 µM, respectively. Thus, in blocking α7 receptors, which are abundantly expressed in the brain, duloxetine exhibited approximately 10-fold to 100- fold higher potency with respect to reported IC50 values for various antidepressant drugs. This may contribute to the antidepressant effect of duloxetine.


Subject(s)
Acetylcholine/pharmacology , Calcium Signaling/drug effects , Chromaffin Cells/drug effects , Duloxetine Hydrochloride/pharmacology , Electrophysiological Phenomena/drug effects , Exocytosis/drug effects , Receptors, Nicotinic/metabolism , Antidepressive Agents/pharmacology , Calcium Channels/metabolism , Catecholamines/metabolism , Chromaffin Cells/cytology , Chromaffin Cells/metabolism , HEK293 Cells , Humans , Nicotinic Antagonists/pharmacology , Sodium Channels/metabolism
6.
J Physiol ; 595(6): 2129-2146, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27982456

ABSTRACT

KEY POINTS: Upon repeated application of short ACh pulses to C57BL6J mouse chromaffin cells, the amperometrically monitored secretory responses promptly decayed to a steady-state level of around 25% of the initial response. A subsequent K+ pulse, however, overcame such decay. These data suggest that mouse chromaffin cells have a ready release-vesicle pool that is selectively recruited by the physiological neurotransmitter ACh. The ACh-sensitive vesicle pool is refilled and maintained by the rate of Ca2+ delivery from mitochondria to the cytosol, through the mitochondrial Na+ /Ca2+ exchanger (mNCX). ITH12662, a novel blocker of the mNCX, prevented the decay of secretion elicited by ACh pulses and delayed the rate of [Ca2+ ]c clearance. This regulatory pathway may be physiologically relevant in situations of prolonged stressful conflicts where a sustained catecholamine release is regulated by mitochondrial Ca2+ circulation through the mNCX, which couples respiration and ATP synthesis to long-term stimulation of chromaffin cells by endogenously released ACh. ABSTRACT: Using caged-Ca2+ photorelease or paired depolarising pulses in voltage-clamped chromaffin cells (CCs), various pools of secretory vesicles with different readiness to undergo exocytosis have been identified. Whether these pools are present in unclamped CCs challenged with ACh, the physiological neurotransmitter at the splanchnic nerve-CC synapse, is unknown. We have explored here whether an ACh-sensitive ready-release vesicle pool (ASP) is present in C57BL6J mouse chromaffin cells (MCCs). Single cells were fast perfused with a Tyrode solution at 37°C, and challenged with 12 sequential ACh pulses (100 µm, 2 s, every 30 s) plus a K+ pulse given at the end (75 mm K+ ). After the first 2-3 ACh pulses the amperometrically monitored secretory responses promptly decayed to a steady-state level of around 25% of the initial response. The last K+ pulse, however, overcame such decay. Repeated ACh pulses to voltage-clamped cells elicited non-desensitising nicotinic currents. Also, the [Ca2+ ]c transients elicited by repeated ACh pulses that were superimposed on a stable baseline elevation did not undergo decay. The novel blocker of the mitochondrial Na+ /Ca2+ exchanger (mNCX) ITH12662 prevented the decay of secretion elicited by ACh pulses and delayed the rate of [Ca2+ ]c clearance. The experiments are compatible with the idea that C57BL6J MCCs have an ASP vesicle pool that is selectively recruited by the physiological neurotransmitter ACh and is regulated by the rate of Ca2+ delivery from mitochondria to the cytosol, through the mNCX.


Subject(s)
Catecholamines/physiology , Chromaffin Cells/physiology , Sodium-Calcium Exchanger/physiology , Acetylcholine/pharmacology , Animals , Calcium/physiology , Cells, Cultured , Chromaffin Cells/drug effects , HeLa Cells , Humans , Male , Mice, Inbred C57BL , Mitochondria/physiology , Potassium/pharmacology
7.
J Neurochem ; 139(5): 722-736, 2016 12.
Article in English | MEDLINE | ID: mdl-27649809

ABSTRACT

Adrenal chromaffin cells (CCs) have been used extensively in studies aimed at revealing the intricacies of the Ca2+ -dependent early and late steps of regulated exocytosis. They have also served as invaluable models to study the kinetics of single-vesicle exocytotic events to infer the characteristics of opening and closing of the exocytotic fusion pore. We have here tested the hypothesis that stimulation at room temperature of CCs from mice C57BL/6 with physiological acetylcholine (ACh) and with other secretagogues (dimethylphenylpiperazinium, high K+ , muscarine, histamine, caffeine), alone or in combination, could trigger amperometric spike events with different kinetics. We found that mean secretory spike events in CCs stimulated with ACh had a fast rise rate of 25 pA/ms and a rapid decay time of 6.2 ms, with a small quantal size (0.31 pC). Surprisingly, these parameters considerably differed from those found in CCs stimulated with all other secretagogues that triggered secretory responses with spike events having smaller rise rates, longer decay times and higher quantal sizes. ACh spikes were unaltered by atropine but mitochondrial protonophore carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone markedly slowed down the rate rise and decay time, and augmented the quantal size of mean secretory events. We conclude that the physiological neurotransmitter ACh triggers a fast and efficient exocytotic response that cannot be mimicked by other secretagogues; such response is regulated by the mitochondrial circulation of calcium ions.


Subject(s)
Acetylcholine/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Catecholamines/metabolism , Chromaffin Cells/drug effects , Chromaffin Cells/metabolism , Animals , Atropine/pharmacology , Caffeine/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Female , Histamine/pharmacology , Kinetics , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...