Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Calcium ; 117: 102819, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37956535

RESUMO

Calcium is a universal intracellular messenger and proper Ca2+concentrations ([Ca2+]) both in the cytosol and in the lumen of cytoplasmic organelles are essential for cell functions. Ca2+ homeostasis is achieved by a delicate pump/leak balance both at the plasma membrane and at the endomembranes, and improper Ca2+ levels result in malfunction and disease. Selective intraorganellar Ca2+measurements are best achieved by using targeted genetically encoded Ca2+ indicators (GECIs) but to calibrate the luminal fluorescent signals into accurate [Ca2+] is challenging, especially in vivo, due to the difficulty to normalize and calibrate the fluorescent signal in various tissues or conditions. We report here a procedure to calibrate the ratiometric signal of GAP (GFP-Aequorin Protein) targeted to the endo-sarcoplasmic reticulum (ER/SR) into [Ca2+]ER/SR based on imaging of fluorescence after heating the tissue at 50-52 °C, since this value coincides with that obtained in the absence of Ca2+ (Rmin). Knowledge of the dynamic range (Rmax/Rmin) and the Ca2+-affinity (KD) of the indicator permits calculation of [Ca2+] by applying a simple algorithm. We have validated this procedure in vitro using several cell types (HeLa, HEK 293T and mouse astrocytes), as well as in vivo in Drosophila. Moreover, this methodology is applicable to other low Ca2+ affinity green and red GECIs.


Assuntos
Equorina , Organelas , Camundongos , Animais , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Calibragem , Organelas/metabolismo , Equorina/metabolismo , Retículo Sarcoplasmático/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio
2.
Cell Metab ; 33(9): 1820-1835.e9, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34343501

RESUMO

Appropriate cristae remodeling is a determinant of mitochondrial function and bioenergetics and thus represents a crucial process for cellular metabolic adaptations. Here, we show that mitochondrial cristae architecture and expression of the master cristae-remodeling protein OPA1 in proopiomelanocortin (POMC) neurons, which are key metabolic sensors implicated in energy balance control, is affected by fluctuations in nutrient availability. Genetic inactivation of OPA1 in POMC neurons causes dramatic alterations in cristae topology, mitochondrial Ca2+ handling, reduction in alpha-melanocyte stimulating hormone (α-MSH) in target areas, hyperphagia, and attenuated white adipose tissue (WAT) lipolysis resulting in obesity. Pharmacological blockade of mitochondrial Ca2+ influx restores α-MSH and the lipolytic program, while improving the metabolic defects of mutant mice. Chemogenetic manipulation of POMC neurons confirms a role in lipolysis control. Our results unveil a novel axis that connects OPA1 in POMC neurons with mitochondrial cristae, Ca2+ homeostasis, and WAT lipolysis in the regulation of energy balance.


Assuntos
Lipólise , Pró-Opiomelanocortina , Tecido Adiposo/metabolismo , Animais , GTP Fosfo-Hidrolases , Homeostase , Camundongos , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo
3.
Pflugers Arch ; 472(4): 439-448, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32246199

RESUMO

Excitability in astroglia is controlled by Ca2+ fluxes from intracellular organelles, mostly from the endoplasmic reticulum (ER). Astrocytic ER possesses inositol 1,4,5-trisphosphate receptors (InsP3R) that can be activated upon stimulation through a vast number of metabotropic G-protein-coupled receptors. By contrast, the role of Ca2+-gated Ca2+ release channels is less explored in astroglia. Here we address this process by monitoring Ca2+ dynamics directly in the cytosol and the ER of astroglial cells. Cultured astrocytes exhibited spontaneous and high-K-evoked cytosolic Ca2+ transients, both of them reversibly abolished by external Ca2+ removal, addition of plasma membrane channel blockers or ER Ca2+ depletion with SERCA inhibitors. Resting astrocyte [Ca2+]ER averaged 400 µM and maximal stimulation with ATP provoked a complete and reversible ER discharge. Direct monitoring of Ca2+ in the lumen of ER showed that high-K induced a Ca2+ release from the ER, and its amplitude was proportional to the [K]. Furthermore, by combining the low affinity GAP3 indicator targeted to the ER with the high affinity cytosolic Rhod-2, we simultaneously imaged ER- and cytosolic-Ca2+ signals, in astrocytes in culture and in situ. Plasma membrane Ca2+ entry triggered a fast ER Ca2+ release coordinated with an increase in cytosolic Ca2+. Thus, we identify a Ca2+-induced Ca2+-release (CICR) mechanism that is likely to participate in spontaneous astroglial oscillations, providing a graded amplification of the cytosolic Ca2+ signal.


Assuntos
Astrócitos/metabolismo , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Membrana Celular/metabolismo , Citosol/metabolismo , Camundongos Endogâmicos C57BL
4.
J Cell Sci ; 133(6)2020 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-32005702

RESUMO

Sarcopenia, the loss of muscle mass and strength associated with age, has been linked to impairment of the cytosolic Ca2+ peak that triggers muscle contraction, but mechanistic details remain unknown. Here we explore the hypothesis that a reduction in sarcoplasmic reticulum (SR) Ca2+ concentration ([Ca2+]SR) is at the origin of this loss of Ca2+ homeostasis. We engineered Drosophila melanogaster to express the Ca2+ indicator GAP3 targeted to muscle SR, and we developed a new method to calibrate the signal into [Ca2+]SRin vivo [Ca2+]SR fell with age from ∼600 µM to 50 µM in close correlation with muscle function, which declined monotonically when [Ca2+]SR was <400 µM. [Ca2+]SR results from the pump-leak steady state at the SR membrane. However, changes in expression of the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump and of the ryanodine receptor leak were too modest to explain the large changes seen in [Ca2+]SR Instead, these changes are compatible with increased leakiness through the ryanodine receptor as the main determinant of the [Ca2+]SR decline in aging muscle. In contrast, there were no changes in endoplasmic reticulum [Ca2+] with age in brain neurons.This article has an associated First Person interview with the first author of the paper.


Assuntos
Cálcio , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Retículo Sarcoplasmático , Animais , Cálcio/metabolismo , Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Retículo Sarcoplasmático/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
5.
Front Endocrinol (Lausanne) ; 11: 615777, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33664709

RESUMO

The adenohypophysis contains five secretory cell types (somatotrophs, lactotrophs, thyrotrophs, corticotrophs, and gonadotrophs), each secreting a different hormone, and controlled by different hypothalamic releasing hormones (HRHs). Exocytic secretion is regulated by cytosolic Ca2+ signals ([Ca2+]C), which can be generated either by Ca2+ entry through the plasma membrane and/or by Ca2+ release from the endoplasmic reticulum (ER). In addition, Ca2+ entry signals can eventually be amplified by ER release via calcium-induced calcium release (CICR). We have investigated the contribution of ER Ca2+ release to the action of physiological agonists in pituitary gland. Changes of [Ca2+] in the ER ([Ca2+]ER) were measured with the genetically encoded low-affinity Ca2+ sensor GAP3 targeted to the ER. We used a transgenic mouse strain that expressed erGAP3 driven by a ubiquitous promoter. Virtually all the pituitary cells were positive for the sensor. In order to mimick the physiological environment, intact pituitary glands or acute slices from the transgenic mouse were used to image [Ca2+]ER. [Ca2+]C was measured simultaneously with Rhod-2. Luteinizing hormone-releasing hormone (LHRH) or thyrotropin releasing hormone (TRH), two agonists known to elicit intracellular Ca2+ mobilization, provoked robust decreases of [Ca2+]ER and concomitant rises of [Ca2+]C. A smaller fraction of cells responded to thyrotropin releasing hormone (TRH). By contrast, depolarization with high K+ triggered a rise of [Ca2+]C without a decrease of [Ca2+]ER, indicating that the calcium-induced calcium-release (CICR) via ryanodine receptor amplification mechanism is not present in these cells. Our results show the potential of transgenic ER Ca2+ indicators as novel tools to explore intraorganellar Ca2+ dynamics in pituitary gland in situ.


Assuntos
Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Imagem Molecular/métodos , Hipófise/citologia , Hipófise/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos
6.
Biochem J ; 475(22): 3639-3649, 2018 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-30389846

RESUMO

Cytosolic Ca2+ signals are often amplified by massive calcium release from the endoplasmic reticulum (ER). This calcium-induced calcium release (CICR) occurs by activation of an ER Ca2+ channel, the ryanodine receptor (RyR), which is facilitated by both cytosolic- and ER Ca2+ levels. Caffeine sensitizes RyR to Ca2+ and promotes ER Ca2+ release at basal cytosolic Ca2+ levels. This outcome is frequently used as a readout for the presence of CICR. By monitoring ER luminal Ca2+ with the low-affinity genetic Ca2+ probe erGAP3, we find here that application of 50 mM caffeine rapidly reduces the Ca2+ content of the ER in HeLa cells by ∼50%. Interestingly, this apparent ER Ca2+ release does not go along with the expected cytosolic Ca2+ increase. These results can be explained by Ca2+ chelation by caffeine inside the ER. Ca2+-overloaded mitochondria also display a drop of the matrix Ca2+ concentration upon caffeine addition. In contrast, in the cytosol, with a low free Ca2+ concentration (10-7 M), no chelation is observed. Expression of RyR3 sensitizes the responses to caffeine with effects both in the ER (increase in Ca2+ release) and in the cytosol (increase in Ca2+ peak) at low caffeine concentrations (0.3-1 mM) that have no effects in control cells. Our results illustrate the fact that simultaneous monitoring of both cytosolic- and ER Ca2+ are necessary to understand the action of caffeine and raise concerns against the use of high concentrations of caffeine as a readout of the presence of CICR.


Assuntos
Cafeína/farmacologia , Cálcio/metabolismo , Citosol/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Citosol/metabolismo , Relação Dose-Resposta a Droga , Retículo Endoplasmático/metabolismo , Células HeLa , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
7.
Cell Calcium ; 64: 3-11, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28214023

RESUMO

Aequorins are excellent tools for measuring intra-organellar Ca2+ and assessing its role in physiological and pathological functions. Here we review targeting strategies to express aequorins in various organelles. We address critical topics such as probe affinity tuning as well as normalization and calibration of the signal. We also focus on bioluminescent Ca2+ imaging in nucleus or mitochondria of living cells. Finally, recent advances with a new chimeric GFP-aequorin protein (GAP), which can be used either as luminescent or fluorescent Ca2+ probe, are presented. GAP is robustly expressed in transgenic flies and mice, where it has proven to be a suitable Ca2+ indicator for monitoring physiological Ca2+ signaling ex vivo and in vivo.


Assuntos
Equorina/metabolismo , Cálcio/metabolismo , Espaço Intracelular/metabolismo , Sondas Moleculares/metabolismo , Organelas/metabolismo , Animais , Corantes Fluorescentes/metabolismo
8.
Biochim Biophys Acta Mol Cell Res ; 1864(6): 894-899, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27939433

RESUMO

GFP-Aequorin Protein (GAP) can be used to measure [Ca2+] inside intracellular organelles, both by luminescence and by fluorescence. The low-affinity variant GAP3 is adequate for ratiometric imaging in the endoplasmic reticulum and Golgi apparatus, and it can be combined with conventional synthetic indicators for simultaneous measurements of cytosolic Ca2+. GAP is bioorthogonal as it does not have mammalian homologues, and it is robust and functionally expressed in transgenic flies and mice, where it can be used for Ca2+ measurements ex vivo and in vivo to explore animal models of health and disease. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.


Assuntos
Equorina/metabolismo , Cálcio/metabolismo , Organelas/metabolismo , Animais , Dípteros , Proteínas de Fluorescência Verde/metabolismo , Humanos , Luminescência , Camundongos , Camundongos Transgênicos
9.
Cell Chem Biol ; 23(6): 738-45, 2016 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-27291400

RESUMO

Proper functioning of organelles such as the ER or the Golgi apparatus requires luminal accumulation of Ca(2+) at high concentrations. Here we describe a ratiometric low-affinity Ca(2+) sensor of the GFP-aequorin protein (GAP) family optimized for measurements in high-Ca(2+) concentration environments. Transgenic animals expressing the ER-targeted sensor allowed monitoring of Ca(2+) signals inside the organelle. The use of the sensor was demonstrated under three experimental paradigms: (1) ER Ca(2+) oscillations in cultured astrocytes, (2) ex vivo functional mapping of cholinergic receptors triggering ER Ca(2+) release in acute hippocampal slices from transgenic mice, and (3) in vivo sarcoplasmic reticulum Ca(2+) dynamics in the muscle of transgenic flies. Our results provide proof of the suitability of the new biosensors to monitor Ca(2+) dynamics inside intracellular organelles under physiological conditions and open an avenue to explore complex Ca(2+) signaling in animal models of health and disease.


Assuntos
Equorina/análise , Cálcio/análise , Cálcio/metabolismo , Proteínas de Fluorescência Verde/análise , Organelas/metabolismo , Equorina/química , Equorina/genética , Equorina/metabolismo , Animais , Drosophila melanogaster , Retículo Endoplasmático/química , Retículo Endoplasmático/metabolismo , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Imagem Molecular , Organelas/química
10.
Cell Calcium ; 58(6): 558-64, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26412347

RESUMO

We have recently described a new class of genetically encoded Ca(2+) indicators composed of two jellyfish proteins, a variant of green fluorescent protein (GFP) and the calcium binding protein apoaequorin, named GAP (Rodriguez-García et al., 2014). GAP is a unique dual-mode Ca(2+) indicator, able to function either as a fluorescent or a luminescent probe, depending on whether the photoprotein aequorin is in its apo-state or reconstituted with its cofactor coelenterazine. We describe here a novel application of GAP as a low affinity bioluminescent indicator, suitable for measurements of [Ca(2+)] in ER or in Golgi apparatus. We used the low affinity variant, GAP1, which carries mutations in two EF-hands of aequorin, reconstituted with coelenterazine n. In comparison to previous bioluminescent aequorin fusions, the decay rate of GAP1 was decreased 8 fold and the affinity for Ca(2+) was lowered one order of magnitude. This improvement allows long-term measurements in high Ca(2+) environments avoiding fast aequorin consumption. GAP1 was targeted to the ER of various cell types, where it monitored resting Ca(2+) concentrations in the range from 400 to 600 µM. ER could be emptied of calcium by stimulation with ATP, carbachol or histamine in intact cells, and by challenge with inositol tris-phosphate in permeabilized cells. GAP1 was also targeted to the Golgi apparatus where it was able to precisely monitor long-term calcium dynamics. GAP1 provides a novel and robust indicator applicable to bioluminescent high-throughput quantitative assays.


Assuntos
Cálcio/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Imidazóis/farmacologia , Luminescência , Substâncias Luminescentes/farmacologia , Organelas/efeitos dos fármacos , Pirazinas/farmacologia , Equorina/metabolismo , Apoproteínas/metabolismo , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Humanos , Organelas/metabolismo , Proteínas Recombinantes/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(7): 2584-9, 2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-24501126

RESUMO

Genetically encoded calcium indicators allow monitoring subcellular Ca(2+) signals inside organelles. Most genetically encoded calcium indicators are fusions of endogenous calcium-binding proteins whose functionality in vivo may be perturbed by competition with cellular partners. We describe here a novel family of fluorescent Ca(2+) sensors based on the fusion of two Aequorea victoria proteins, GFP and apo-aequorin (GAP). GAP exhibited a unique combination of features: dual-excitation ratiometric imaging, high dynamic range, good signal-to-noise ratio, insensitivity to pH and Mg(2+), tunable Ca(2+) affinity, uncomplicated calibration, and targetability to five distinct organelles. Moreover, transgenic mice for endoplasmic reticulum-targeted GAP exhibited a robust long-term expression that correlated well with its reproducible performance in various neural tissues. This biosensor fills a gap in the actual repertoire of Ca(2+) indicators for organelles and becomes a valuable tool for in vivo Ca(2+) imaging applications.


Assuntos
Equorina/metabolismo , Técnicas Biossensoriais/métodos , Cálcio/análise , Imagem Molecular/métodos , Organelas/química , Equorina/genética , Animais , Retículo Endoplasmático/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos
12.
PLoS One ; 7(7): e41530, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22848520

RESUMO

Besides regulating energy balance and reducing body-weight, the adipokine leptin has been recently shown to be neuroprotective and antiapoptotic by promoting neuronal survival after excitotoxic and oxidative insults. Here, we investigated the firing properties of mouse hippocampal neurons and the effects of leptin pretreatment on hypoxic damage (2 hours, 3% O(2)). Experiments were carried out by means of the microelectrode array (MEA) technology, monitoring hippocampal neurons activity from 11 to 18 days in vitro (DIV). Under normoxic conditions, hippocampal neurons were spontaneously firing, either with prevailing isolated and randomly distributed spikes (11 DIV), or with patterns characterized by synchronized bursts (18 DIV). Exposure to hypoxia severely impaired the spontaneous activity of hippocampal neurons, reducing their firing frequency by 54% and 69%, at 11 and 18 DIV respectively, and synchronized their firing activity. Pretreatment with 50 nM leptin reduced the firing frequency of normoxic neurons and contrasted the hypoxia-induced depressive action, either by limiting the firing frequency reduction (at both ages) or by increasing it to 126% (in younger neurons). In order to find out whether leptin exerts its effect by activating large conductance Ca(2+)-activated K(+) channels (BK), as shown on rat hippocampal neurons, we applied the BK channel blocker paxilline (1 µM). Our data show that paxilline reversed the effects of leptin, both on normoxic and hypoxic neurons, suggesting that the adipokine counteracts hypoxia through BK channels activation in mouse hippocampal neurons.


Assuntos
Hipocampo/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Leptina/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Animais , Hipóxia Celular/efeitos dos fármacos , Células Cultivadas , Hipocampo/citologia , Indóis/farmacologia , Camundongos , Bloqueadores dos Canais de Potássio/farmacologia
13.
Gen Physiol Biophys ; 30 Spec No: S44-53, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21869451

RESUMO

L-type calcium channels (LTCC) are important functional elements of hippocampal neurons contributing to processes like memory formation and gene expression. Mice lacking the Ca(V)1.2 channel in hippocampal pyramidal cells exhibited defects in spatial memory (Moosmang et al. 2005) and lowered frequency of repetitive action potential (AP) firing (Lacinova et al. 2008). We tested the contribution of LTCC to AP firing of cultured rat neonatal hippocampal neurons using the dihydropyridine channel blocker nimodipine. Ionic currents and APs were recorded in the whole cell patch clamp configuration. A prolonged depolarizing current pulse activated the firing of a series of APs. The presence of 10 µM nimodipine blocked all but the first AP in series. This concentration, which is potent enough to completely block LTCC, inhibited about 35-50% of the total calcium current. In addition, nimodipine blocked about 50% of both calcium-dependent and voltage-dependent potassium currents whereas the sodium current was not affected. We suggest that nimodipine suppressed the firing of APs in cultured neonatal rat hippocampal neurons due to inhibition of both calcium and potassium currents.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Hipocampo/citologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Nimodipina/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Bário/metabolismo , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Condutividade Elétrica , Neurônios/metabolismo , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA