Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(15)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34360957

ABSTRACT

In recent years, the "non-autonomous motor neuron death" hypothesis has become more consolidated behind amyotrophic lateral sclerosis (ALS). It postulates that cells other than motor neurons participate in the pathology. In fact, the involvement of the autonomic nervous system is fundamental since patients die of sudden death when they become unable to compensate for cardiorespiratory arrest. Mitochondria are thought to play a fundamental role in the physiopathology of ALS, as they are compromised in multiple ALS models in different cell types, and it also occurs in other neurodegenerative diseases. Our study aimed to uncover mitochondrial alterations in the sympathoadrenal system of a mouse model of ALS, from a structural, bioenergetic and functional perspective during disease instauration. We studied the adrenal chromaffin cell from mutant SOD1G93A mouse at pre-symptomatic and symptomatic stages. The mitochondrial accumulation of the mutated SOD1G93A protein and the down-regulation of optic atrophy protein-1 (OPA1) provoke mitochondrial ultrastructure alterations prior to the onset of clinical symptoms. These changes affect mitochondrial fusion dynamics, triggering mitochondrial maturation impairment and cristae swelling, with increased size of cristae junctions. The functional consequences are a loss of mitochondrial membrane potential and changes in the bioenergetics profile, with reduced maximal respiration and spare respiratory capacity of mitochondria, as well as enhanced production of reactive oxygen species. This study identifies mitochondrial dynamics regulator OPA1 as an interesting therapeutic target in ALS. Additionally, our findings in the adrenal medulla gland from presymptomatic stages highlight the relevance of sympathetic impairment in this disease. Specifically, we show new SOD1G93A toxicity pathways affecting cellular energy metabolism in non-motor neurons, which offer a possible link between cell specific metabolic phenotype and the progression of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , GTP Phosphohydrolases/metabolism , Mitochondria/metabolism , Superoxide Dismutase-1/genetics , Adrenal Glands/cytology , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cells, Cultured , Chromaffin Cells/metabolism , Down-Regulation , GTP Phosphohydrolases/genetics , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mitochondria/ultrastructure , Mutation, Missense , Reactive Oxygen Species/metabolism , Superoxide Dismutase-1/metabolism
2.
Cell Signal ; 76: 109811, 2020 12.
Article in English | MEDLINE | ID: mdl-33075487

ABSTRACT

Resveratrol (RESV) is one of the most abundant polyphenol-stilbene compounds found in red wine with well-established cardioprotective and antihypertensive effects. Hyperactivity of the sympathoadrenal axis seems to be one of the major contributing factors in the pathogenesis of human essential hypertension. Alterations in outward voltage-dependent potassium currents (IK) and inward voltage-dependent sodium (INa), calcium (ICa) and nicotinic (IACh) currents, CCs excitability, Ca2+ homeostasis, and catecholamine exocytosis were previously related to the hypertensive state. This raised the issue of whether in vivo long-term RESV treatment can directly act as a modulator of Ca2+ influx or a regulator of ion channel permeability in CCs. We monitored outward and inward currents, and cytosolic Ca2+ concentrations ([Ca2+]c) using different pharmacological approaches in CCs from normotensive (WKY) and hypertensive (SHR) animals chronically exposed to trans-RESV (50 mg/L/v.o, 28 days). The long-term RESV treatment prevented the increase of the systolic blood pressure (SBP) in SHR, without reversion of cardiac hypertrophy. We also found an increase of the outward IK, reduction in inward INa,ICa, and IACh, and the mitigation of [Ca2+]c overload in CCs from SHR at the end of RESV treatment. Our data revealed that electrophysiological alterations of the CCs and in its Ca2+ homeostasis are potential new targets related to the antihypertensive effects of long-term RESV treatment.


Subject(s)
Antihypertensive Agents/pharmacology , Chromaffin Cells/drug effects , Hypertension/drug therapy , Resveratrol/pharmacology , Animals , Calcium/metabolism , Chromaffin Cells/pathology , Male , Primary Cell Culture , Rats , Rats, Inbred SHR , Rats, Inbred WKY
3.
Med Res Rev ; 40(6): 2427-2465, 2020 11.
Article in English | MEDLINE | ID: mdl-32677086

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Pharmaceutical Preparations , Animals , Humans , Mice , Neurodegenerative Diseases/drug therapy , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7
4.
J Neurochem ; 152(3): 299-314, 2020 02.
Article in English | MEDLINE | ID: mdl-31677273

ABSTRACT

Adrenal chromaffin cells release epinephrine (EPI) and norepinephrine (NE) into the bloodstream as part of the homeostatic response to situations like stress. Here we utilized EPI-deficient mice generated by knocking out (KO) the phenylethanolamine N-methyltransferase (Pnmt) gene. These Pnmt-KO mice were bred to homozygosis but displayed no major phenotype. The lack of EPI was partially compensated by an increase in NE, suggesting that EPI storage was optimized in adrenergic cells. Electron microscopy showed that despite the lack of EPI, chromaffin granules retain their shape and general appearance. This indicate that granules from adrenergic or noradrenergic cells preserve their characteristics even though they contain only NE. Acute insulin injection largely reduced the EPI content in wild-type animals, with a minimal reduction in NE, whereas there was only a partial reduction in NE content in Pnmt-KO mice. The analysis of exocytosis by amperometry revealed a reduction in the quantum size (-30%) and Imax (-21%) of granules in KO cells relative to the wild-type granules, indicating a lower affinity of NE for the granule matrix of adrenergic cells. As amperometry cannot distinguish between adrenergic or noradrenergic cells, it would suggest even a larger reduction in the affinity for the matrix. Therefore, our results demonstrate that adrenergic cells retain their structural characteristics despite the almost complete absence of EPI. Furthermore, the chromaffin granule matrix from adrenergic cells is optimized to accumulate EPI, with NE being a poor substitute. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.


Subject(s)
Chromaffin Cells/metabolism , Chromaffin Granules/metabolism , Epinephrine/metabolism , Norepinephrine/metabolism , Animals , Exocytosis/physiology , Male , Mice , Mice, Knockout , Phenylethanolamine N-Methyltransferase/deficiency , Phenylethanolamine N-Methyltransferase/genetics
5.
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
6.
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
7.
Pflugers Arch ; 470(8): 1255-1270, 2018 08.
Article in English | MEDLINE | ID: mdl-29721607

ABSTRACT

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.


Subject(s)
Calcium/metabolism , Chromaffin Cells/drug effects , Exocytosis/drug effects , Hydrogen Sulfide/pharmacology , Animals , Calcium Channels/metabolism , Calcium Signaling/drug effects , Catecholamines/metabolism , Cattle , Cells, Cultured , Chromaffin Cells/metabolism , Cytosol/drug effects , Cytosol/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Morpholines/pharmacology , Organothiophosphorus Compounds/pharmacology , Potassium/metabolism
8.
Eur J Pharmacol ; 815: 416-426, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28970016

ABSTRACT

Comorbidity of diabetes and hypertension is frequent. Here, we have performed a comparative study in three animal models namely, normotensive Wistar Kyoto (WKY) rats, streptozotocin-induced diabetic rats (STZ), and spontaneously hypertensive rats (SHR). With respect WKY rats, we have found the following alterations in adrenal chromaffin cells from STZ and SHR rats: (1) diminished Ca2+ currents; (2) augmented [Ca2+]c elevations and catecholamine release in cells stimulated with angiotensin II or high K+; (3) unchanged expression of angiotensin II receptors AT1 and AT2; (4) higher density of secretory vesicles at subplasmalemmal sites; (5) mitochondria with lower cristae density that were partially depolarized; and (6) lower whole cell ATP content. These alterations may have their origin in (i) an augmented capacity of the endoplasmic reticulum [Ca2+] store likely due to (ii) impaired mitochondrial Ca2+ uptake; (iii) augmented high-[Ca2+]c microdomains at subplasmalemmal sites secondary to augmented calcium-induce calcium release and to inositol tris-phosphate receptor mediated enhanced Ca2+ mobilization from the endoplasmic reticulum; and (iv) augmented vesicle pool. These alterations seem to be common to the two models of human hypertension here explored, STZ diabetic rats and SHR hypertensive rats.


Subject(s)
Calcium Signaling , Catecholamines/metabolism , Chromaffin Cells/metabolism , Chromaffin Cells/pathology , Diabetes Mellitus, Experimental/pathology , Mitochondria/pathology , Animals , Cell Count , Male , Rats , Rats, Inbred SHR , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism
9.
Oxid Med Cell Longev ; 2017: 1682393, 2017.
Article in English | MEDLINE | ID: mdl-28588746

ABSTRACT

Studies have reported the importance of mitochondria in sperm functionality. However, for some species, the glycolytic pathway appears to be as important as oxidative phosphorylation in ATP synthesis and sperm kinetics. These mechanisms have not been fully elucidated for bovine spermatozoa. Therefore, the aim of this study was to evaluate the role of mitochondria and the glycolytic pathway in ATP synthesis, sperm movement patterns, and oxidative homeostasis of epididymal spermatozoa in bovine specimens. We observed that mitochondrial uncoupling with protonophores significantly reduced ATP levels. However, these levels were reestablished after stimulation of the glycolytic pathway. We verified the same pattern of results for sperm kinetic variables and the production of reactive oxygen species (ROS). Thus, we suggest that, after its appropriate stimulation, the glycolytic pathway is capable of maintaining ATP levels, sperm kinetic patterns, and oxidative balance of bovine epididymal spermatozoa submitted to mitochondrial uncoupling.


Subject(s)
Adenosine Triphosphate/metabolism , Glycolysis/drug effects , Reactive Oxygen Species/metabolism , Spermatozoa/metabolism , Uncoupling Agents/pharmacology , Animals , Cattle , Epididymis/metabolism , Male , Spermatozoa/cytology
10.
J Hypertens ; 35(8): 1594-1608, 2017 08.
Article in English | MEDLINE | ID: mdl-28403042

ABSTRACT

OBJECTIVE: It has been suggested that Ca entry through store-operated Ca channels (SOCs) is regulated by a dynamic interplay between the endoplasmic reticulum Ca stores and the mitochondria. These relationships drive the activation and inactivation of SOCs, yet it remains unclear whether this regulation of SOCs by mitochondria is altered in the aorta of spontaneously hypertensive rats (SHRs). METHODS: We performed a thorough study of the mitochondrial membrane potential, the ability of mitochondria to deal with cytosolic Ca, capacitative Ca entry (CCE), and stromal interaction molecule 1 (STIM1) and calcium release-activated calcium modulator 1 (orai1) protein expression, as well as the contractile capacity of aortic rings, in normotensive Wistar Kyoto rats (WKYs) and SHRs. RESULTS: Changes were observed in aortic tissue and cultured vascular smooth muscle cells isolated from SHRs relative to WKYs, including more depolarized mitochondria, stronger CCE upon the addition of Ca, larger cytosolic Ca transients (cytosolic Ca concentration) or aortic ring contraction elicited by endoplasmic reticulum depletion and a significant increase in STIM1 protein expression but not of orai1. CONCLUSION: These results suggest that the impaired Ca buffering capacity of partially depolarized mitochondria dysregulates CCE, leading to overfilling of the endoplasmic reticulum Ca store through enhanced STIM1/orai1 interactions and an increase in aorta contractions in SHRs. Thus, understanding the implications of the alterations to STIM1/orai1, and their relationship to mitochondria, may aid drug development and therapeutic strategies to treat hypertension, as well as its long-term sequelae in poorly controlled patients.


Subject(s)
Aorta/physiopathology , Calcium Channels/metabolism , Calcium/metabolism , Hypertension/physiopathology , Animals , Aorta/metabolism , Endoplasmic Reticulum/metabolism , Male , Mitochondria/metabolism , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY
11.
Eur J Pharmacol ; 803: 65-77, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28322841

ABSTRACT

It is known that chronic ethanol (EtOH) consumption leads to hypertension development and has been associated with deleterious effects on the cardiovascular system. Whether this condition alters calcium (Ca2+) signaling and exocytosis in adrenal chromaffin cells (CCs) as the case is for genetic hypertension, is unknown. We explored this question in four randomized experimental groups, male Wistar Kyoto (WKY/EtOH) and Spontaneously Hypertensive (SHR/EtOH) rats were subjected to the intake of increasing EtOH concentrations (5-20%, for 30 days) and their respective controls (WKY/Control and SHR/Control) received water. WKY/EtOH developed hypertension and cardiac hypertrophy; blood aldehyde dehydrogenase (ALDH) and H2O2 were also augmented. In comparison with WKY/Control, CCs from WKY/EtOH had the following features: (i) depolarization and higher frequency of spontaneous action potentials; (ii) decreased Ca2+ currents with slower inactivation; (iii) decreased K+ currents; (iv) augmented K+-elicited cytosolic Ca2+ transients ([Ca2+]c); (v) enhanced K+-elicited catecholamine release. These cardiovascular, blood and CCs changes were qualitatively similar to those undergone by SHR/Control and SHR/EtOH. The results suggest that the hypertension elicited by chronic EtOH has pathogenic features common to genetic hypertension namely, augmented [Ca2+]c transients and catecholamine release from their CCs.


Subject(s)
Alcohol Drinking/adverse effects , Catecholamines/metabolism , Chromaffin Cells/drug effects , Chromaffin Cells/metabolism , Electrophysiological Phenomena/drug effects , Hypertension/chemically induced , Hypertension/pathology , Action Potentials/drug effects , Animals , Calcium/metabolism , Chromaffin Cells/pathology , Cytosol/drug effects , Cytosol/metabolism , Ethanol/pharmacology , Hypertension/metabolism , Hypertension/physiopathology , Male , Potassium/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Time Factors
12.
Curr Vasc Pharmacol ; 15(3): 265-281, 2017.
Article in English | MEDLINE | ID: mdl-28155613

ABSTRACT

BACKGROUND: Chronic ethanol (EtOH) consumption has been associated with deleterious effects on the cardiovascular system by abnormal calcium (Ca2+) handling. Store-operated Ca2+ entry (SOCE) is related to cardiovascular remodeling which leads to the hypertension development, and the coupling between STIM-1 (ER Ca2+ sensor) and Orai-1 (channel pore) is a key mechanism to control SOCE through of store-operated Ca2+ channels (SOCCs). However, the role of STIM-1/Orai-1-mediated SOCE and its cross-talk with EtOH-triggered vascular remodeling and hypertension remain poorly understood. We address this subject in the present study by evaluating how chronic EtOH consumption induces alterations in Ca2+ handling via SOCE. METHODS: Male Wistar Kyoto (WKY) and Spontaneously Hypertensive (SHR) rats were subjected to the intake of increasing EtOH concentrations (5-20%, for 30 days). Systolic blood pressure (SBP) and EtOH concentration were measured; cardiovascular remodeling was assessed by histomorphometry; and function/ expression of STIM-1/Orai-1-mediated Ca2+ influx were evaluated by isometric contraction and western blot experiments. RESULTS: Compared to the WKY-Control, our results show that: (1) chronic EtOH consumption caused a significant elevation of SBP in both strains; (2) cardiac hypertrophy and hypertrophic aortic wall remodeling much more pronounced in WKY-EtOH; (3) decreased capacity of ER to store and release Ca2+; (4) increased STIM-1/Orai-1-mediated SOCCs activation, which was selectively inhibited by YM-58483; and (5) increased expression of STIM-1 in WKY-EtOH and SHR-Control rats. CONCLUSION: These findings suggest that hypertrophic aortic remodeling and abnormal contraction triggered mainly by Ca2+ overload via STIM-1/Orai-1-mediated SOCE through SOCCs are involved hypertension developed by EtOH consumption.


Subject(s)
Alcohol Drinking/adverse effects , Calcium Signaling , Calcium/metabolism , Ethanol , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Blood Pressure , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Hypertension/etiology , Hypertension/physiopathology , Male , Muscle, Smooth, Vascular/physiopathology , Myocytes, Cardiac/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Time Factors , Up-Regulation , Vascular Remodeling , Vasoconstriction
14.
ACS Chem Neurosci ; 8(1): 67-81, 2017 01 18.
Article in English | MEDLINE | ID: mdl-27731633

ABSTRACT

During brain ischemia, oxygen and glucose deprivation induces calcium overload, extensive oxidative stress, neuroinflammation, and, finally, massive neuronal loss. In the search of a neuroprotective compound to mitigate this neuronal loss, we have designed and synthesized a new multitarget hybrid (ITH14001) directed at the reduction of calcium overload by acting on two regulators of calcium homeostasis; the mitochondrial Na+/Ca2+ exchanger (mNCX) and L-type voltage dependent calcium channels (VDCCs). This compound is a hybrid of CGP37157 (mNCX inhibitor) and nimodipine (L-type VDCCs blocker), and its pharmacological evaluation revealed a moderate ability to selectively inhibit both targets. These activities conferred concentration-dependent neuroprotection in two models of Ca2+ overload, such as toxicity induced by high K+ in the SH-SY5Y cell line (60% protection at 30 µM) and veratridine in hippocampal slices (26% protection at 10 µM). It also showed neuroprotective effect against oxidative stress, an activity related to its nitrogen radical scavenger effect and moderate induction of the Nrf2-ARE pathway. Its Nrf2 induction capability was confirmed by the increase of the expression of the antioxidant and anti-inflammatory enzyme heme-oxygenase I (3-fold increase). In addition, the multitarget profile of ITH14001 led to anti-inflammatory properties, shown by the reduction of nitrites production induced by lipopolysaccharide in glial cultures. Finally, it showed protective effect in two acute models of cerebral ischemia in hippocampal slices, excitotoxicity induced by glutamate (31% protection at 10 µM) and oxygen and glucose deprivation (76% protection at 10 µM), reducing oxidative stress and iNOS deleterious induction. In conclusion, our hybrid derivative showed improved neuroprotective properties when compared to its parent compounds CGP37157 and nimodipine.


Subject(s)
Brain Ischemia/drug therapy , Calcium/metabolism , Nimodipine/pharmacology , Nimodipine/therapeutic use , Oxidative Stress/drug effects , Thiazepines/therapeutic use , Animals , Animals, Newborn , Benzodiazepinones/chemistry , Benzodiazepinones/pharmacology , Benzodiazepinones/therapeutic use , Brain Ischemia/pathology , Cattle , Cell Hypoxia/drug effects , Cell Line, Tumor , Cells, Cultured , Chromaffin Cells , Disease Models, Animal , Embryo, Mammalian , Hippocampus/drug effects , Hippocampus/pathology , Male , Neuroblastoma/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nimodipine/analogs & derivatives , Nimodipine/chemistry , Rats , Rats, Sprague-Dawley , Thiazepines/chemistry , Thiazepines/pharmacology
15.
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
16.
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
17.
Pflugers Arch ; 467(12): 2589-601, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26254593

ABSTRACT

Calcium buffering by mitochondria plays a relevant physiological function in the regulation of Ca(2+) and exocytotic signals in mature chromaffin cells (CCs) from various adult mammals. Whether a similar or different role of mitochondrial Ca(2+) buffering is present in immature CCs at early life has not been explored. Here we present a comparative study in rat embryonic CCs and rat mother CCs, of various physiological parameters that are known to be affected by mitochondrial Ca(2+) buffering during cell activation. We found that the clearance of cytosolic Ca(2+) transients ([Ca(2+)]c) elicited by high K(+) was 7-fold faster in embryo CCs compared to mother CCs. This strongly suggests that at embryonic life, the mitochondria play a more significant role in the clearance of [Ca(2+)]c loads compared to adult life. Consistent with this view are the following results concerning the transient suppression of mitochondrial Ca(2+) buffering by protonophore FCCP, in embryonic CCs compared to mother CCs: (i) faster and greater inactivation of inward calcium currents, (ii) higher K(+)-elicited [Ca(2+)]c transients with 25-fold faster clearance, (iii) higher increase of basal catecholamine release and (iv) higher potentiation of K(+)-evoked secretion. These pronounced differences could be explained by two additional features (embryo versus mother CCs): (a) slower recovery of mitochondrial resting membrane potential after the application of a transient FCCP pulse and (b) greater relative density of the mitochondria in the cytosol. This tighter control by the mitochondria of Ca(2+) and exocytotic signals may be relevant to secure a healthy catecholamine secretory response at early life.


Subject(s)
Calcium Signaling , Chromaffin Cells/metabolism , Exocytosis , Adrenal Medulla/cytology , Adrenal Medulla/embryology , Adrenal Medulla/metabolism , Animals , Catecholamines/pharmacology , Cells, Cultured , Chromaffin Cells/drug effects , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...