Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurochem ; 158(2): 153-168, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33704788

RESUMO

γ-Aminobutyric acid (GABA) is thought to play a paracrine role in adrenal medullary chromaffin (AMC) cells. Comparative physiological and immunocytochemical approaches were used to address the issue of how the paracrine function of GABA in AMC cells is established. GABAA receptor Cl- channel activities in AMC cells of rats and mice, where corticosterone is the major glucocorticoid, were much smaller than those in AMC cells of guinea-pigs and cattle, where cortisol is the major. The extent of enhancement of GABAA receptor α3 subunit expression in rat pheochromocytoma (PC12) cells by cortisol was larger than that by corticosterone in parallel with their glucocorticoid activities. Thus, the species difference in GABAA receptor expression may be ascribed to a difference in glucocorticoid activity between corticosterone and cortisol. GABAA receptor Cl- channel activity in mouse AMC cells was enhanced by allopregnanolone, as noted with that in guinea-pig AMC cells, and the enzymes involved in allopregnanolone production were immunohistochemically detected in the zona fasciculata in both mice and guinea pigs. The expression of glutamic acid decarboxylase 67 (GAD67), one of the GABA synthesizing enzymes, increased after birth, whereas GABAA receptors already developed at birth. Stimulation of pituitary adenylate cyclase-activating polypeptide (PACAP) receptors, but not nicotinic or muscarinic receptors, in PC12 cells, resulted in an increase in GAD67 expression in a protein-kinase A-dependent manner. The results indicate that glucocorticoid and PACAP are mainly responsible for the expressions of GABAA receptors and GAD67 involved in GABA signaling in AMC cells, respectively.


Assuntos
Medula Suprarrenal/fisiologia , Células Cromafins/fisiologia , Comunicação Parácrina/fisiologia , Ácido gama-Aminobutírico/fisiologia , Medula Suprarrenal/citologia , Animais , Bovinos , Canais de Cloreto/metabolismo , Cricetinae , Glutamato Descarboxilase/metabolismo , Cobaias , Hidrocortisona/metabolismo , Imuno-Histoquímica , Masculino , Mesocricetus , Camundongos , Camundongos Endogâmicos C57BL , Células PC12 , Pregnanolona/farmacologia , Ratos , Receptores de GABA-A/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/efeitos dos fármacos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo
2.
JCI Insight ; 6(2)2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33491666

RESUMO

Reduced expression of the plasma membrane citrate transporter INDY (acronym I'm Not Dead, Yet) extends life span in lower organisms. Deletion of the mammalian Indy (mIndy) gene in rodents improves metabolism via mechanisms akin to caloric restriction, known to lower blood pressure (BP) by sympathoadrenal inhibition. We hypothesized that mIndy deletion attenuates sympathoadrenal support of BP. Continuous arterial BP and heart rate (HR) were reduced in mINDY-KO mice. Concomitantly, urinary catecholamine content was lower, and the decreases in BP and HR by mIndy deletion were attenuated after autonomic ganglionic blockade. Catecholamine biosynthesis pathways were reduced in mINDY-KO adrenals using unbiased microarray analysis. Citrate, the main mINDY substrate, increased catecholamine content in pheochromocytoma cells, while pharmacological inhibition of citrate uptake blunted the effect. Our data suggest that deletion of mIndy reduces sympathoadrenal support of BP and HR by attenuating catecholamine biosynthesis. Deletion of mIndy recapitulates beneficial cardiovascular and metabolic responses to caloric restriction, making it an attractive therapeutic target.


Assuntos
Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Transportadores de Ácidos Dicarboxílicos/genética , Transportadores de Ácidos Dicarboxílicos/fisiologia , Sistema Simpático-Suprarrenal/fisiologia , Simportadores/genética , Simportadores/fisiologia , Glândulas Suprarrenais/anatomia & histologia , Glândulas Suprarrenais/fisiologia , Animais , Restrição Calórica , Catecolaminas/biossíntese , Linhagem Celular , Células Cromafins/fisiologia , Transportadores de Ácidos Dicarboxílicos/deficiência , Expressão Gênica , Frequência Cardíaca/genética , Frequência Cardíaca/fisiologia , Longevidade/genética , Longevidade/fisiologia , Malatos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Cardiovasculares , Atividade Motora/genética , Atividade Motora/fisiologia , Piridinas/farmacologia , Simportadores/deficiência
3.
Compr Physiol ; 9(4): 1443-1502, 2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31688964

RESUMO

Chromaffin cells (CCs) of the adrenal gland and the sympathetic nervous system produce the catecholamines (epinephrine and norepinephrine; EPI and NE) needed to coordinate the bodily "fight-or-flight" response to fear, stress, exercise, or conflict. EPI and NE release from CCs is regulated both neurogenically by splanchnic nerve fibers and nonneurogenically by hormones (histamine, corticosteroids, angiotensin, and others) and paracrine messengers [EPI, NE, adenosine triphosphate, opioids, γ-aminobutyric acid (GABA), etc.]. The "stimulus-secretion" coupling of CCs is a Ca2+ -dependent process regulated by Ca2+ entry through voltage-gated Ca2+ channels, Ca2+ pumps, and exchangers and intracellular organelles (RE and mitochondria) and diffusible buffers that provide both Ca2+ -homeostasis and Ca2+ -signaling that ultimately trigger exocytosis. CCs also express Na+ and K+ channels and ionotropic (nAChR and GABAA ) and metabotropic receptors (mACh, PACAP, ß-AR, 5-HT, histamine, angiotensin, and others) that make CCs excitable and responsive to autocrine and paracrine stimuli. To maintain high rates of E/NE secretion during stressful conditions, CCs possess a large number of secretory chromaffin granules (CGs) and members of the soluble NSF-attachment receptor complex protein family that allow docking, fusion, and exocytosis of CGs at the cell membrane, and their recycling. This article attempts to provide an updated account of well-established features of the molecular processes regulating CC function, and a survey of the as-yet-unsolved but important questions relating to CC function and dysfunction that have been the subject of intense research over the past 15 years. Examples of CCs as a model system to understand the molecular mechanisms associated with neurodegenerative diseases are also provided. Published 2019. Compr Physiol 9:1443-1502, 2019.


Assuntos
Doenças das Glândulas Suprarrenais/patologia , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/fisiologia , Células Cromafins/fisiologia , Doenças das Glândulas Suprarrenais/metabolismo , Regulação da Expressão Gênica , Humanos , Sistema Nervoso Simpático/citologia , Sistema Nervoso Simpático/fisiologia
4.
ACS Chem Neurosci ; 10(5): 2459-2466, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30892011

RESUMO

Adenosine triphosphate (ATP) is the main energy source for cellular metabolism. Besides that, ATP is a neurotransmitter and a cotransmitter that acts on purinergic receptors present either pre- or postsynaptically. Almost all types of secretory vesicles from any neuron or animal species contain high concentrations of ATP, being an essential factor in the accumulation of neurotransmitters. In this work, we studied the effects of ATP on quantum catecholamine release and vesicular storage in chromaffin cells. We combined three electrochemical methods: conventional amperometry with intracellular vesicle impact electrochemical cytometry and vesicle impact electrochemical cytometry. We found that extracellular ATP increased the released quantal fraction of catecholamine but not its vesicular content. Studying the dynamics of exocytosis events in ATP treated cells showed that ATP affects the release fusion pore. To elucidate the mechanisms of the observed ATP effects, cells and vesicles were pharmacologically treated with suramin (a purinergic blocker) and ARL-67156 (an antagonist of ecto-ATPases). The data indicate that the catecholamine content of vesicles increased compared to control after these drugs. Our data suggest that ATP acting on purinergic receptors increases the quantum releasable size through an increased fusion pore opening and that ARL-67156 and/or suramin protect the vesicle from neurotransmitter leakage by functioning as competitive inhibitors to ATP.


Assuntos
Trifosfato de Adenosina/fisiologia , Células Cromafins/fisiologia , Exocitose/fisiologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Catecolaminas/metabolismo , Bovinos , Células Cultivadas , Agonistas Purinérgicos/farmacologia , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P2/metabolismo , Suramina/farmacologia
5.
Pflugers Arch ; 470(1): 79-88, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28965274

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) was first identified in hypothalamus, based on its ability to elevate cyclic AMP in the anterior pituitary. PACAP has been identified as the adrenomedullary neurotransmitter in stress through a combination of ex vivo, in vivo, and in cellula experiments over the past two decades. PACAP causes catecholamine secretion, and activation of catecholamine biosynthetic enzymes, during episodes of stress in mammals. Features of PACAP signaling allowing stress transduction at the splanchnicoadrenomedullary synapse have yielded insights into the contrasting roles of acetylcholine's and PACAP's actions as first messengers at the chromaffin cell, via differential release at low and high rates of splanchnic nerve firing, and differential signaling pathway engagement leading to catecholamine secretion and chromaffin cell gene transcription. Secretion stimulated by PACAP, via calcium influx independent of action potential generation, is under active investigation in several laboratories both at the chromaffin cell and within autonomic ganglia of both the parasympathetic and sympathetic nervous systems. PACAP is a neurotransmitter important in stress transduction in the central nervous system as well, and is found at stress-transduction nuclei in brain including the paraventricular nucleus of hypothalamus, the amygdala and extended amygdalar nuclei, and the prefrontal cortex. The current status of PACAP as a master regulator of stress signaling in the nervous system derives fundamentally from the establishment of its role as the splanchnicoadrenomedullary transmitter in stress. Experimental elucidation of PACAP action at this synapse remains at the forefront of understanding PACAP's role in stress signaling throughout the nervous system.


Assuntos
Células Cromafins/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Estresse Fisiológico , Transmissão Sináptica , Animais , Catecolaminas/metabolismo , Células Cromafins/fisiologia , Humanos
6.
Pflugers Arch ; 470(1): 21-27, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29058146

RESUMO

During the last 10 years, we have been working on human chromaffin cells obtained from the adrenal gland of organ donors that suffered encephalic or cardiac death. We first electrophysiologically characterized the nicotinic acetylcholine receptors (nAChRs) activated by acetylcholine, and their contribution to the exocytosis of chromaffin vesicles and release of catecholamines. We have shown that these cells possess an adrenergic phenotype. This phenotype may contribute to an increased expression of α7 nAChRs in these cells, allowing for recording of α7 nAChR currents, something that had previously not been achieved in non-human species. The use of α-conotoxins allowed us to characterize non-α7 nAChR subtypes and, together with molecular biology experiments, conclude that the predominant nAChR subtype in human chromaffin cells is α3ß4* (asterisk indicates the posible presence of additional subunits). In addition, there is a minor population of αxß2 nAChRs. Both α7 and non-α7 nAChR subtypes contribute to the exocytotic process. Exocytosis mediated by nAChRs could be as large in magnitude as that elicited by calcium entry through voltage-dependent calcium channels. Finally, we have also investigated the effect of nAChR-targeted tobacco cessation drugs on catecholamine release in chromaffin cells. We have concluded that at therapeutic concentrations, varenicline alone does not increase the frequency of action potentials evoked by ACh. However, varenicline in the presence of nicotine does increase this frequency, and thus, in the presence of both drugs, the probability of increased catecholamine release in human chromaffin cells is high.


Assuntos
Células Cromafins/metabolismo , Conotoxinas/farmacologia , Neurotoxinas/farmacologia , Agonistas Nicotínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/metabolismo , Potenciais de Ação , Sinalização do Cálcio , Células Cromafins/efeitos dos fármacos , Células Cromafins/fisiologia , Humanos , Receptores Nicotínicos/química
7.
J Neurochem ; 140(1): 37-52, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27805736

RESUMO

Varenicline is a nicotinic acetylcholine receptor (nAChR) agonist used to treat nicotine addiction, but a live debate persists concerning its mechanism of action in reducing nicotine consumption. Although initially reported as α4ß2 selective, varenicline was subsequently shown to activate other nAChR subtypes implicated in nicotine addiction including α3ß4. However, it remains unclear whether activation of α3ß4 nAChRs by therapeutically relevant concentrations of varenicline is sufficient to affect the behavior of cells that express this subtype. We used patch-clamp electrophysiology to assess the effects of varenicline on native α3ß4* nAChRs (asterisk denotes the possible presence of other subunits) expressed in human adrenal chromaffin cells and compared its effects to those of nicotine. Varenicline and nicotine activated α3ß4* nAChRs with EC50 values of 1.8 (1.2-2.7) µM and 19.4 (11.1-33.9) µM, respectively. Stimulation of adrenal chromaffin cells with 10 ms pulses of 300 µM acetylcholine (ACh) in current-clamp mode evoked sodium channel-dependent action potentials (APs). Under these conditions, perfusion of 50 or 100 nM varenicline showed very little effect on AP firing compared to control conditions (ACh stimulation alone), but at higher concentrations (250 nM) varenicline increased the number of APs fired up to 436 ± 150%. These results demonstrate that therapeutic concentrations of varenicline are unlikely to alter AP firing in chromaffin cells. In contrast, nicotine showed no effect on AP firing at any of the concentrations tested (50, 100, 250, and 500 nM). However, perfusion of 50 nM nicotine simultaneously with 100 nM varenicline increased AP firing by 290 ± 104% indicating that exposure to varenicline and nicotine concurrently may alter cellular behavior such as excitability and neurotransmitter release.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Córtex Suprarrenal/efeitos dos fármacos , Células Cromafins/efeitos dos fármacos , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Vareniclina/administração & dosagem , Potenciais de Ação/fisiologia , Córtex Suprarrenal/citologia , Córtex Suprarrenal/fisiologia , Adulto , Idoso , Animais , Células Cromafins/fisiologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Xenopus laevis
8.
J Cell Biol ; 210(5): 785-800, 2015 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-26323692

RESUMO

Annexin A2, a calcium-, actin-, and lipid-binding protein involved in exocytosis, mediates the formation of lipid microdomains required for the structural and spatial organization of fusion sites at the plasma membrane. To understand how annexin A2 promotes this membrane remodeling, the involvement of cortical actin filaments in lipid domain organization was investigated. 3D electron tomography showed that cortical actin bundled by annexin A2 connected docked secretory granules to the plasma membrane and contributed to the formation of GM1-enriched lipid microdomains at the exocytotic sites in chromaffin cells. When an annexin A2 mutant with impaired actin filament-bundling activity was expressed, the formation of plasma membrane lipid microdomains and the number of exocytotic events were decreased and the fusion kinetics were slower, whereas the pharmacological activation of the intrinsic actin-bundling activity of endogenous annexin A2 had the opposite effects. Thus, annexin A2-induced actin bundling is apparently essential for generating active exocytotic sites.


Assuntos
Anexina A2/metabolismo , Membrana Celular/metabolismo , Células Cromafins/fisiologia , Exocitose/fisiologia , Vesículas Secretórias/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Anexina A2/genética , Catecolaminas/metabolismo , Bovinos , Células Cultivadas , Tomografia com Microscopia Eletrônica , Fusão de Membrana/fisiologia , Microdomínios da Membrana/metabolismo , Nicotina/farmacologia , Estrutura Terciária de Proteína , beta-Galactosidase/metabolismo
9.
ACS Chem Neurosci ; 6(9): 1626-36, 2015 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-26192029

RESUMO

Mitochondria regulate cellular Ca(2+) oscillations, taking up Ca(2+) through its uniporter and releasing it through the mitochondrial sodium/calcium exchanger. The role of mitochondria in the regulation of Ca(2+) cycle has received much attention recently, as it is a central stage in neuronal survival and death processes. Over the last decades, the 4,1-benzothiazepine CGP37157 has been the only available blocker of the mitochondrial sodium/calcium exchanger, although it targets several other calcium transporters. We report the synthesis of 4,1-benzothiazepine derivatives with the goal of enhancing mitochondrial sodium/calcium exchanger blockade and selectivity, and the evaluation of their cytoprotective effect. The compound 4c presented an interesting neuroprotective profile in addition to an important blockade of the mitochondrial sodium/calcium exchanger. The use of this benzothiazepine could help to understand the physiological functions of the mitochondrial sodium/calcium exchanger. In addition, we hypothesize that a moderate blockade of the mitochondrial sodium/calcium exchanger would provide enhanced neuroprotection in neurons.


Assuntos
Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Tiazepinas/farmacologia , Animais , Cálcio/metabolismo , Bovinos , Morte Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cromafins/efeitos dos fármacos , Células Cromafins/fisiologia , Citoproteção , Avaliação Pré-Clínica de Medicamentos , Células HeLa , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estrutura Molecular , Fármacos Neuroprotetores/síntese química , Fármacos Neuroprotetores/química , Sódio/metabolismo , Trocador de Sódio e Cálcio/antagonistas & inibidores , Trocador de Sódio e Cálcio/metabolismo , Tiazepinas/síntese química , Tiazepinas/química
10.
Neuroscience ; 301: 134-43, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26047729

RESUMO

The effects of mitochondrial inhibitors (CN(-), a complex IV inhibitor and CCCP, protonophore) on catecholamine (CA) secretion and mitochondrial function were explored functionally and biochemically in rat and guinea-pig adrenal chromaffin cells. Guinea-pig chromaffin cells conspicuously secreted CA in response to CN(-) or CCCP, but rat cells showed a little, if any, secretory response to either of them. The resting metabolic rates in rat adrenal medullae did not differ from those in guinea-pig adrenal medullae. On the other hand, the time course of depolarization of the mitochondrial membrane potential (ΔΨm) in guinea-pig chromaffin cells in response to CN(-) was slower than that in rat chromaffin cells, and this difference was abolished by oligomycin, an F1F0-ATPase inhibitor. The extent of CCCP-induced decrease in cellular ATP in guinea-pig chromaffin cells, which was indirectly measured using a Mg(2+) indicator, was smaller than that in rat chromaffin cells. Relative expression levels of F1F0-ATPase inhibitor factor in guinea-pig adrenal medullae were smaller than in rat adrenal medullae, and the opposite was true for F1F0-ATPase α subunit. The present results indicate that guinea-pig chromaffin cells secrete more CA in response to a mitochondrial inhibitor than rat chromaffin cells and this higher susceptibility in the former is accounted for by a larger extent of reversed operation of F1F0-ATPase with the consequent decrease in ATP under conditions where ΔΨm is depolarized.


Assuntos
Medula Suprarrenal/fisiologia , Catecolaminas/metabolismo , Células Cromafins/fisiologia , Mitocôndrias/fisiologia , Consumo de Oxigênio/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Medula Suprarrenal/efeitos dos fármacos , Animais , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Hipóxia Celular , Células Cromafins/efeitos dos fármacos , Cobaias , Mitocôndrias/efeitos dos fármacos , Fosforilação Oxidativa , Ratos , Ratos Wistar , Especificidade da Espécie
11.
Philos Trans R Soc Lond B Biol Sci ; 370(1672)2015 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-26009767

RESUMO

Little is known about the interactions between nicotinic and muscarinic acetylcholine receptors (nAChRs and mAChRs). Here we report that methacholine (MCh), a selective agonist of mAChRs, inhibited up to 80% of nicotine-induced nAChR currents in sympathetic superior cervical ganglion neurons and adrenal chromaffin cells. The muscarine-induced inhibition (MiI) substantially reduced ACh-induced membrane currents through nAChRs and quantal neurotransmitter release. The MiI was time- and temperature-dependent. The slow recovery of nAChR current after washout of MCh, as well as the high value of Q10 (3.2), suggested, instead of a direct open-channel blockade, an intracellular metabotropic process. The effects of GTP-γ-S, GDP-ß-S and pertussis toxin suggested that MiI was mediated by G-protein signalling. Inhibitors of protein kinase C (bisindolymaleimide-Bis), protein kinase A (H89) and PIP2 depletion attenuated the MiI, indicating that a second messenger pathway is involved in this process. Taken together, these data suggest that mAChRs negatively modulated nAChRs via a G-protein-mediated second messenger pathway. The time dependence suggests that MiI may provide a novel mechanism for post-synaptic adaptation in all cells/neurons and synapses expressing both types of AChRs.


Assuntos
Células Cromafins/fisiologia , Cloreto de Metacolina/farmacologia , Neurônios/fisiologia , Antagonistas Nicotínicos/farmacologia , Gânglio Cervical Superior/citologia , Transmissão Sináptica/fisiologia , Animais , Células Cromafins/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas de Ligação ao GTP/metabolismo , Agonistas Muscarínicos/farmacologia , Neurônios/metabolismo , Técnicas de Patch-Clamp , Proteína Quinase C/antagonistas & inibidores , Ratos , Sistemas do Segundo Mensageiro/fisiologia , Gânglio Cervical Superior/fisiologia , Temperatura , Fatores de Tempo
12.
Nat Commun ; 6: 6297, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25708831

RESUMO

In neurosecretory cells, secretory vesicles (SVs) undergo Ca(2+)-dependent fusion with the plasma membrane to release neurotransmitters. How SVs cross the dense mesh of the cortical actin network to reach the plasma membrane remains unclear. Here we reveal that, in bovine chromaffin cells, SVs embedded in the cortical actin network undergo a highly synchronized transition towards the plasma membrane and Munc18-1-dependent docking in response to secretagogues. This movement coincides with a translocation of the cortical actin network in the same direction. Both effects are abolished by the knockdown or the pharmacological inhibition of myosin II, suggesting changes in actomyosin-generated forces across the cell cortex. Indeed, we report a reduction in cortical actin network tension elicited on secretagogue stimulation that is sensitive to myosin II inhibition. We reveal that the cortical actin network acts as a 'casting net' that undergoes activity-dependent relaxation, thereby driving tethered SVs towards the plasma membrane where they undergo Munc18-1-dependent docking.


Assuntos
Actinas/metabolismo , Células Cromafins/fisiologia , Proteínas Munc18/metabolismo , Miosina Tipo II/metabolismo , Neurossecreção , Vesículas Secretórias/fisiologia , Animais , Bovinos , Compostos Heterocíclicos de 4 ou mais Anéis , Células PC12 , Ratos
13.
J Neurosci ; 35(4): 1380-9, 2015 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-25632116

RESUMO

Activity-dependent bulk endocytosis allows neurons to internalize large portions of the plasma membrane in response to stimulation. However, whether this critical type of compensatory endocytosis is unique to neurons or also occurs in other excitable cells is currently unknown. Here we used fluorescent 70 kDa dextran to demonstrate that secretagogue-induced bulk endocytosis also occurs in bovine chromaffin cells. The relatively large size of the bulk endosomes found in this model allowed us to investigate how the neck of the budding endosomes constricts to allow efficient recruitment of the fission machinery. Using time-lapse imaging of Lifeact-GFP-transfected chromaffin cells in combination with fluorescent 70 kDa dextran, we detected acto-myosin II rings surrounding dextran-positive budding endosomes. Importantly, these rings were transient and contracted before disappearing, suggesting that they might be involved in restricting the size of the budding endosome neck. Based on the complete recovery of dextran fluorescence after photobleaching, we demonstrated that the actin ring-associated budding endosomes were still connected with the extracellular fluid. In contrast, no such recovery was observed following the constriction and disappearance of the actin rings, suggesting that these structures were pinched-off endosomes. Finally, we showed that the rings were initiated by a circular array of phosphatidylinositol(4,5)bisphosphate microdomains, and that their constriction was sensitive to both myosin II and dynamin inhibition. The acto-myosin II rings therefore play a key role in constricting the neck of budding bulk endosomes before dynamin-dependent fission from the plasma membrane of neurosecretory cells.


Assuntos
Actinas/metabolismo , Células Cromafins/fisiologia , Células Cromafins/ultraestrutura , Endocitose/fisiologia , Endossomos/metabolismo , Miosina Tipo II/metabolismo , Glândulas Suprarrenais/citologia , Animais , Transporte Biológico/efeitos dos fármacos , Bovinos , Membrana Celular/metabolismo , Células Cultivadas , Células Cromafins/efeitos dos fármacos , Dextranos/metabolismo , Dinaminas/antagonistas & inibidores , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Endossomos/ultraestrutura , Glicosilfosfatidilinositóis/genética , Glicosilfosfatidilinositóis/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Hidrazonas/farmacologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Miosina Tipo II/antagonistas & inibidores , Naftóis/farmacologia , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Rodaminas/metabolismo , Fatores de Tempo , Transfecção
14.
Biointerphases ; 9(1): 011003, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24739010

RESUMO

Neuronal process growth is guided by extrinsic environmental cues such as extracellular matrix (ECM) proteins. Recent reports have described that the growth cone extension is superior across gradients of the ECM protein laminin compared to growth across uniformly distributed laminin. In this work, the authors have prepared gradients of laminin on aligned electrospun nanofibers for use as substrates for neuronal growth. The substrates therefore presented both topographical and chemical guidance cues. Step gradients were prepared by the controlled robotic immersion of plasma-treated polycaprolactone fibers reacted with N-hydroxysuccinimide into the protein solution. The gradients were analyzed using x-ray photoelectron spectroscopy and confocal laser scanning microscopy. Gradients with a dynamic range of protein concentrations were successfully generated and neurite outgrowth was evaluated using neuronlike pheochromocytoma cell line 12 (PC12) cells. After 10 days of culture, PC12 neurite lengths varied from 32.7 ± 14.2 µm to 76.3 ± 9.1 µm across the protein concentration gradient. Neurite lengths at the highest concentration end of the gradient were significantly longer than neurite lengths observed for cells cultured on samples with uniform protein coverage. Gradients were prepared both in the fiber direction and transverse to the fiber direction. Neurites preferentially aligned with the fiber direction in both cases indicating that fiber alignment has a more dominant role in controlling neurite orientation, compared to the chemical gradient.


Assuntos
Células Cromafins/efeitos dos fármacos , Células Cromafins/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Laminina/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Animais , Microscopia Confocal , Células PC12 , Espectroscopia Fotoeletrônica , Ratos
15.
J Alzheimers Dis ; 41(1): 129-49, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24595198

RESUMO

Pyroglutamate amyloid-ß peptides (pGlu-Aß) are particularly pernicious forms of amyloid-ß peptides (Aß) present in Alzheimer's disease (AD) brains. pGlu-Aß peptides are N-terminally truncated forms of full-length Aß peptides (flAß(1-40/42)) in which the N-terminal glutamate is cyclized to pyroglutamate to generate pGlu-Aß(3-40/42). ß-secretase cleavage of amyloid-ß precursor protein (AßPP) produces flAß(1-40/42), but it is not yet known whether the ß-secretase BACE1 or the alternative ß-secretase cathepsin B (CatB) participate in the production of pGlu-Aß. Therefore, this study examined the effects of gene knockout of these proteases on brain pGlu-Aß levels in transgenic AßPPLon mice, which express AßPP isoform 695 and have the wild-type (wt) ß-secretase activity found in most AD patients. Knockout or overexpression of the CatB gene reduced or increased, respectively, pGlu-Aß(3-40/42), flAß(1-40/42), and pGlu-Aß plaque load, but knockout of the BACE1 gene had no effect on those parameters in the transgenic mice. Treatment of AßPPLon mice with E64d, a cysteine protease inhibitor of CatB, also reduced brain pGlu-Aß(3-42), flAß(1-40/42), and pGlu-Aß plaque load. Treatment of neuronal-like chromaffin cells with CA074Me, an inhibitor of CatB, resulted in reduced levels of pGlu-Aß(3-40) released from the activity-dependent, regulated secretory pathway. Moreover, CatB knockout and E64d treatment has been previously shown to improve memory deficits in the AßPPLon mice. These data illustrate the role of CatB in producing pGlu-Aß and flAß that participate as key factors in the development of AD. The advantages of CatB inhibitors, especially E64d and its derivatives, as alternatives to BACE1 inhibitors in treating AD patients are discussed.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Catepsina B/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Leucina/análogos & derivados , Ácido Pirrolidonocarboxílico/análise , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/patologia , Catepsina B/antagonistas & inibidores , Catepsina B/genética , Bovinos , Células Cultivadas , Células Cromafins/efeitos dos fármacos , Células Cromafins/fisiologia , Dipeptídeos/farmacologia , Humanos , Leucina/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/tratamento farmacológico , Placa Amiloide/patologia , Placa Amiloide/fisiopatologia
16.
J Clin Endocrinol Metab ; 98(11): 4346-54, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24001749

RESUMO

CONTEXT: A number of incidentally discovered pheochromocytomas are not associated with hypertension. The characteristics of normotensive incidentally discovered pheochromocytomas (NIPs) are poorly known. OBJECTIVE: The purpose of this work was to assess the clinical, hormonal, histological, and molecular features of NIPs. DESIGN: This was a retrospective cohort recruited from 2001 to 2011 in 2 tertiary care medical departments. PATIENTS AND METHODS: Clinical, biological, and radiological investigations performed in 96 consecutive patients with sporadic unilateral pheochromocytomas were examined; 47 patients had overt pheochromocytomas responsible for hypertension. Among the patients with incidental pheochromocytomas, 28 had hypertension and 21 were normotensive (NIPs). A total of 62 tumors were examined to determine the Pheochromocytoma of the Adrenal Gland Scale Score, and 29 were studied for the expression of 16 genes involved in chromaffin cell function. RESULTS: Tumor size and metaiodobenzylguanidine (MIBG) scintigraphy results were similar for hypertensive pheochromocytomas (HPs) and NIPs. Patients with NIPs displayed reduced summed levels of urinary catecholamines and metanephrines and, more specifically, reduced levels of adrenaline and metadrenaline compared with those of patients with HPs (P < .001). Urinary metanephrines had 98% diagnostic sensitivity in patients with HPs and only 75% in patients with NIPs (P < .01). Tumor diameter positively correlated with the total amount of urinary concentrations of metanephrines in patients with HPs (P < .001) but not in patients with NIPs. NIPs displayed global decreased chromaffin gene expression (reaching significance for 5 of them) and 2 corresponding proteins (phenylethanolamine N-methyltransferase and secretogranin II) and a significant increase in the cellularity, mitotic activity, and presence of atypical mitosis (P < .05). CONCLUSIONS: NIPs differ from pheochromocytomas responsible for hypertension and display features of altered chromaffin differentiation. These tumors may be misdiagnosed with the use of the usual biological diagnostic tools.


Assuntos
Neoplasias das Glândulas Suprarrenais , Células Cromafins/diagnóstico por imagem , Células Cromafins/fisiologia , Regulação Neoplásica da Expressão Gênica , Feocromocitoma , 3-Iodobenzilguanidina , Neoplasias das Glândulas Suprarrenais/diagnóstico por imagem , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/metabolismo , Células Cromafins/patologia , Feminino , Humanos , Hipertensão/diagnóstico por imagem , Hipertensão/genética , Hipertensão/metabolismo , Achados Incidentais , Masculino , Pessoa de Meia-Idade , Feocromocitoma/diagnóstico por imagem , Feocromocitoma/genética , Feocromocitoma/metabolismo , Cintilografia , Compostos Radiofarmacêuticos , Estudos Retrospectivos , Transcriptoma
17.
Eur J Pharmacol ; 706(1-3): 25-35, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23499685

RESUMO

Enhanced activity of the sympatho-adrenal axis and augmented circulating catecholamines has been implicated in the development of hypertension. Release of catecholamine from stimulated adrenal medulla chromaffin cells has been shown to be higher and longer in spontaneously hypertensive rats (SHRs), compared with normotensive Wistar rats (NWRs). Whether differences in the functional expression of voltage-dependent calcium channels (VDCCs) of the L-, N-, or P/Q subtypes may contribute to such distinct secretory behaviour, is unknown. We therefore approached here this study in voltage-clamped NWR and SHR chromaffin cells, using 10mM Ba(2+) as charge carrier (IBa) and selective blockers of each channel type. We found that compared with NWR cells, SHR chromaffin cells exhibited the following differences: (1) 30% diminution of the IBa fraction carried by L channels; (2) a doubling of the IBa fraction carried by P/Q channels; (3) more visible current modulation by ATP that could be linked to a 10-fold higher mRNA levels for purinergic receptors of the P2Y2 subtype; and (3) a higher contribution of PQ channels to the transients of the cytosolic calcium concentrations ([Ca(2+)]c) generated by K(+), compared with L channels. These results may contribute to the better understanding of the greater calcium signalling and exocytotic responses of SHR compared with NWR chromaffin cells, found in three previous reports from our laboratories.


Assuntos
Medula Suprarrenal/fisiologia , Canais de Cálcio/fisiologia , Células Cromafins/fisiologia , Hipertensão/metabolismo , Trifosfato de Adenosina/farmacologia , Medula Suprarrenal/citologia , Medula Suprarrenal/efeitos dos fármacos , Animais , Bário/farmacologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Hipertensão/fisiopatologia , Técnicas In Vitro , Masculino , Potássio/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Wistar , Receptores Purinérgicos P2Y2/genética
18.
J Physiol ; 590(20): 5053-73, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22826131

RESUMO

Mouse chromaffin cells (MCCs) express high densities of L-type Ca2+ channels (LTCCs), which control pacemaking activity and catecholamine secretion proportionally to their density of expression. In vivo phosphorylation of LTCCs by cAMP-PKA and cGMP­PKG, regulate LTCC gating in two opposing ways: the cAMP-PKA pathway potentiates while the cGMP­PKG cascade inhibits LTCCs. Despite this, no attempts have been made to answer three key questions related to the two Cav1 isoforms expressed in MCCs (Cav1.2 and Cav1.3): (i) how much are the two Cav1 channels basally modulated by PKA and PKG?, (ii) to what extent can Cav1.2 and Cav1.3 be further regulated by PKA or PKG activation?, and (iii) are the effects of both kinases cumulative when simultaneously active? Here, by comparing the size of L-type currents of wild-type (WT; Cav1.2+Cav1.3) and Cav1.3−/− KO (Cav1.2) MCCs, we provide new evidence that both PKA and PKG pathways affect Cav1.2 and Cav1.3 to the same extent either under basal conditions or induced stimulation. Inhibition of PKA by H89 (5 µM) reduced the L-type current in WT and KO MCCs by∼60%,while inhibition of PKG by KT 5823 (1 µM) increased by∼40% the same current in both cell types. Given that Cav1.2 and Cav1.3 carry the same quantity of Ca2+ currents, this suggests equal sensitivity of Cav1.2 and Cav1.3 to the two basal modulatory pathways. Maximal stimulation of cAMP­PKA by forskolin (100 µM) and activation of cGMP­PKG by pCPT-cGMP (1mM) uncovered a∼25% increase of L-type currents in the first case and∼65% inhibition in the second case in both WT and KO MCCs, suggesting equal sensitivity of Cav1.2 and Cav1.3 during maximal PKA or PKG stimulation. The effects of PKA and PKG were cumulative and most evident when one pathway was activated and the other was inhibited. The two extreme combinations(PKA activation­PKG inhibition vs. PKG activation-PKA inhibition) varied the size of L-type currents by one order of magnitude (from 180% to 18% of control size). Taken together our data suggest that: (i) Cav1.2 and Cav1.3 are equally sensitive to PKA and PKG action under both basal conditions and maximal stimulation, and (ii) PKA and PKG act independently on both Cav1.2 and Cav1.3, producing cumulative effects when opposingly activated. These extreme Cav1 channel modulations may occur either during high-frequency sympathetic stimulation to sustain prolonged catecholamine release (maximal L-type current) or following activation of the NO­cGMP­PKG signalling pathway (minimal L-type current) to limit the steady release of catecholamines.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Animais , Células Cultivadas , Células Cromafins/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
J Mol Neurosci ; 48(2): 403-12, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22610912

RESUMO

It has been known for more than a decade that the neuropeptide PACAP (pituitary adenylate cyclase-activating polypeptide) is co-stored with acetylcholine in the splanchnic nerve terminals innervating the adrenal medulla. Both transmitters are robust secretagogues for catecholamine release from chromaffin cells. Here, we review the unique contribution of PACAP to the functioning of the splanchnic-adrenal synapse in stress. While acetylcholine is released across a wide range of firing frequencies, PACAP is released only at high frequencies of stimulation, and its role in the regulation of epinephrine secretion and biosynthesis is highly specialized. PACAP is responsible for long-term catecholamine secretion using secretory mechanisms different from the rapidly desensitizing depolarization evoked by acetylcholine through nicotinic receptor activation. PACAP signaling also maintains catecholamine synthesis required for sustained secretion during prolonged stress via induction of the enzymes TH and PNMT, and enhances transcription of additional secreted molecules found in chromaffin cells that alter further secretion through both autocrine and paracrine mechanisms. PACAP thus mediates chromaffin cell plasticity via functional encoding of cellular experience. These features of PACAP action at the splanchnic-adrenal synapse may be paradigmatic for the general actions of neuropeptides as effectors of stimulus-secretion-synthesis coupling in stress.


Assuntos
Medula Suprarrenal/fisiologia , Células Cromafins/fisiologia , Neurotransmissores/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Transdução de Sinais/fisiologia , Estresse Fisiológico/fisiologia , Sinapses/fisiologia , Medula Suprarrenal/citologia , Medula Suprarrenal/metabolismo , Animais , Catecolaminas/metabolismo , Células Cromafins/citologia , Células Cromafins/metabolismo , Humanos , Neurotransmissores/metabolismo
20.
Am J Physiol Cell Physiol ; 303(3): C257-66, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22592408

RESUMO

Neuroendocrine adrenal medullary chromaffin cells receive synaptic excitation through the sympathetic splanchnic nerve to elicit catecholamine release into the circulation. Under basal sympathetic tone, splanchnic-released acetylcholine evokes chromaffin cells to fire action potentials, leading to synchronous phasic catecholamine release. Under elevated splanchnic firing, experienced under the sympathoadrenal stress response, chromaffin cells undergo desensitization to cholinergic excitation. Yet, stress evokes a persistent and elevated adrenal catecholamine release. This sustained stress-evoked release has been shown to depend on splanchnic release of a peptide transmitter, pituitary adenylate cyclase-activating peptide (PACAP). PACAP stimulates catecholamine release through a PKC-dependent pathway that is mechanistically independent of cholinergic excitation. Moreover, it has also been reported that shorter term phospho-regulation of existing gap junction channels acts to increase junctional conductance. In this study, we test if PACAP-mediated excitation upregulates cell-cell electrical coupling to enhance chromaffin cell excitability. We utilize electrophysiological recordings conducted in adrenal tissue slices to measure the effects of PACAP stimulation on cell coupling. We report that PACAP excitation increases electrical coupling and the spread of electrical excitation between adrenal chromaffin cells. Thus PACAP acts not only as a secretagogue but also evokes an electrical remodeling of the medulla, presumably to adapt to the organism's needs during acute sympathetic stress.


Assuntos
Medula Suprarrenal/efeitos dos fármacos , Junções Comunicantes/efeitos dos fármacos , Neurotransmissores/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Medula Suprarrenal/fisiologia , Animais , Células Cromafins/efeitos dos fármacos , Células Cromafins/fisiologia , Fenômenos Eletrofisiológicos , Feminino , Junções Comunicantes/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA